AC IMMUNE
SA
TABLE OF CONTENTS
|
1
|
|
|
2
|
|
2
|
|
A.
|
|
2
|
|
B.
|
|
2
|
|
C.
|
|
2
|
|
2
|
|
A.
|
|
2
|
|
B.
|
|
2
|
|
2
|
|
A.
|
|
3
|
|
B.
|
|
3
|
|
C.
|
|
3
|
|
D.
|
|
3
|
|
48
|
|
A.
|
|
48
|
|
B.
|
|
48
|
|
C.
|
|
113
|
|
D.
|
|
114
|
|
114
|
|
114
|
|
A.
|
|
114
|
|
B.
|
|
130
|
|
C.
|
|
132
|
|
D.
|
|
133
|
|
E.
|
|
133
|
|
133
|
|
A.
|
|
133
|
|
B.
|
|
137
|
|
C.
|
|
139
|
|
D.
|
|
142
|
|
E.
|
|
142
|
|
142
|
|
A.
|
|
142
|
|
B.
|
|
144
|
|
C.
|
|
144
|
|
144
|
|
A.
|
|
144
|
|
B.
|
|
145
|
|
145
|
|
A.
|
|
145
|
|
B.
|
|
145
|
|
C.
|
|
145
|
|
D.
|
|
145
|
|
E.
|
|
145
|
|
F.
|
|
145
|
|
145
|
|
A.
|
|
145
|
|
B.
|
|
145
|
|
C.
|
|
145
|
|
D.
|
|
145
|
|
E.
|
|
146
|
|
F.
|
|
152
|
|
G.
|
|
152
|
|
H.
|
|
153
|
|
I.
|
|
153
|
|
153
|
|
154
|
|
A.
|
|
154
|
|
B.
|
|
154
|
|
C.
|
|
154
|
|
D.
|
|
154
|
|
|
154
|
|
154
|
|
A.
|
|
154
|
|
B.
|
|
155
|
|
155
|
|
155
|
|
A.
|
|
155
|
|
B.
|
|
155
|
|
C.
|
|
155
|
|
D.
|
|
155
|
|
155
|
|
155
|
|
156
|
|
156
|
|
156
|
|
156
|
|
156
|
|
156
|
|
157
|
|
157
|
|
157
|
|
157
|
|
158
|
|
158
|
PRESENTATION OF
FINANCIAL AND OTHER INFORMATION
Unless otherwise indicated or the
context otherwise requires, all references in this annual report on
Form 20-F (the “Annual Report”) to “AC Immune,” “ACIU,” “Company,”
“we,” “our,” “ours,” “us” or similar terms refer to AC Immune SA
together with its subsidiary. The Company owns various registered
and unregistered trademarks, for some of which protection has been
obtained or is being sought, including Morphomer™, SupraAntigen™
and its corporate name, logo and Nasdaq Global Market symbol. All
other trademarks, trade names and service marks of other companies
appearing in this Annual Report are the property of their
respective owners. Solely for convenience, the trademarks and trade
names in this Annual Report may be referred to without the ™
symbols, but such references should not be construed as any
indicator that their respective owners will not assert, to the
fullest extent under applicable law, their rights thereto. The
Company does not intend to use or display other companies’
trademarks and/or trade names to imply a relationship with, or
endorsement or sponsorship of the Company by, any other
companies.
Financial statements
Our consolidated financial
statements are presented in Swiss Francs and in accordance with
International Financial Reporting Standards (IFRS), as issued by
the International Accounting Standards Board (IASB). None of the
consolidated financial statements was prepared in accordance with
generally accepted accounting principles in the United States (US).
The terms “dollar” and “USD” refer to US dollars, and the terms
“Swiss Franc” and “CHF” refer to the legal currency of Switzerland,
unless otherwise indicated. We have made rounding adjustments to
some of the figures included in this Annual Report. Accordingly,
any numerical discrepancies in any table between totals and sums of
the amounts listed are due to rounding.
FORWARD-LOOKING STATEMENTS
This Annual Report contains
statements that constitute forward-looking statements. All
statements other than statements of historical facts contained in
this Annual Report, including statements regarding our future
results of operations and financial position, business strategy,
product candidates, product pipeline, ongoing and planned clinical
studies, including those of our collaboration partners, regulatory
approvals, research and development (R&D) costs, timing and
likelihood of success, as well as plans and objectives of
management for future operations, are forward-looking statements.
Many of the forward-looking statements contained in this Annual
Report can be identified by the use of forward-looking words such
as “anticipate,” “believe,” “could,” “expect,” “should,” “plan,”
“intend,” “estimate,” “will” and “potential,” among others.
Forward-looking statements appear
in a number of places in this Annual Report and include, but are
not limited to, statements regarding our intent, belief or current
expectations. Forward-looking statements are based on our
management’s beliefs and assumptions, and on information currently
available to our management. Such statements are subject to risks
and uncertainties, and actual results may differ materially from
those expressed or implied in the forward-looking statements due to
various factors, including, but not limited to, those identified
under “Item 3. Key information—D. Risk factors” in this Annual
Report. These risks and uncertainties include multiple
factors:
|
• |
the success of our and our collaboration partners’ clinical
studies, and our and their ability to obtain and maintain
regulatory approval and to commercialize ACI-35, semorinemab,
ACI-7104, Morphomer Tau, ACI-24 for Alzheimer’s disease (AD) and
for Down syndrome-related AD (ACI-24 for DS), crenezumab, PI-2620,
our Tau-positron emission tomography (PET) imaging tracer, our
alpha-synuclein (a-syn) PET tracer and to a lesser extent our
preclinical candidates;
|
|
• |
the preclinical and clinical safety, efficacy and utility of
our product candidates;
|
|
• |
the ability of our competitors to discover, develop or
commercialize competing products before or more successfully than
we do;
|
|
• |
our plans to research, develop and commercialize our product
candidates;
|
|
• |
the identification of serious adverse, undesirable or
unacceptable side effects related to our product candidates;
|
|
• |
our ability to maintain our current strategic relationships
with our collaboration partners;
|
|
• |
our ability to protect and maintain our, and not infringe on
third parties’, intellectual property rights throughout the
world;
|
|
• |
our ability to raise capital when needed in order to continue
our product development programs or commercialization
efforts;
|
|
• |
our ability to attract and retain qualified employees and key
personnel;
|
|
• |
the acceptance by the Food and Drug Administration (FDA) and
applicable foreign regulatory authorities of data from studies that
we and our collaboration partners conduct within and outside the US
now and in the future;
|
|
• |
our foreign private issuer status, the loss of which would
require us to comply with the Exchange Act’s domestic reporting
regime, and cause us to incur significant legal, accounting and
other expenses;
|
|
• |
our incorporation in Switzerland, the laws of which govern our
corporate affairs and may differ from those applicable to companies
incorporated in the US; and
|
|
• |
the other risk factors discussed under “Item 3. Key
information—D. Risk factors.”
|
These forward-looking statements
are applicable only as of the date of this Annual Report, and are
subject to a number of risks, uncertainties and assumptions
described under the sections in this Annual Report entitled “Item
3. Key information—D. Risk factors” and “Item 5. Operating and
financial review and prospects,” and elsewhere in this Annual
Report. Because forward-looking statements are inherently subject
to risks and uncertainties, some of which cannot be predicted or
quantified and some of which are beyond our control, you should not
rely on these forward-looking statements as predictions of future
events. The events and circumstances reflected in our
forward-looking statements may not be achieved or occur and actual
results could differ materially from those projected in the
forward-looking statements. Moreover, we operate in an evolving
environment. New risk factors and uncertainties may emerge from
time to time, and it is not possible for management to predict all
risk factors and uncertainties. Except as required by applicable
law, we do not plan to publicly update or revise any
forward-looking statements contained herein, whether as a result of
any new information, future events, changed circumstances or
otherwise.
ITEM 1.
|
IDENTITY OF
DIRECTORS, SENIOR MANAGEMENT AND ADVISERS
|
A.
|
Directors and senior management
|
Not applicable.
Not applicable.
Not applicable.
ITEM 2.
|
OFFER STATISTICS
AND EXPECTED TIMETABLE
|
Not applicable.
B.
|
Method and expected timetable
|
Not applicable.
B.
|
Capitalization and indebtedness
|
Not applicable.
C.
|
Reasons for the offer and use of
proceeds
|
Not applicable.
You should
carefully consider the risks and uncertainties described below and
the other information in this Annual Report before making an
investment in our common shares. Our business, financial condition
or results of operations could be materially and adversely affected
if any of these risks occurs, and as a result, the market price of
our common shares could decline and you could lose all or part of
your investment. This Annual Report also contains forward-looking
statements that involve risks and uncertainties. See
“Forward-Looking Statements.” Our actual results could differ
materially and adversely from those anticipated in these
forward-looking statements as a result of certain factors.
The below
provides a summary of our principal risk factors:
Risks
related to our business:
|
• |
We depend heavily on the
success of our clinical and, to a lesser extent, preclinical
products:
|
|
a. |
Our ability to generate
product revenues, which we do not expect to occur for several
years, will depend on clinical and regulatory success which have
low probabilities of success in the central nervous system (CNS)
space in which we operate.
|
|
• |
Results of early preclinical
and clinical studies may not be predictive of future results:
|
|
a. |
Products that show positive or timely preclinical or early
clinical results may not show sufficient safety or efficacy in
later-stage clinical studies and therefore may fail to obtain
regulatory approvals.
|
|
• |
Our products may not gain market acceptance or may be
preempted by competitors:
|
|
a. |
Even if our products obtain regulatory approval, they may not
be accepted by healthcare providers, patients or the medical
community.
|
|
b. |
Our success is dependent on
the ability to discover, develop and obtain marketing approval for
our products. We face and will continue to face intense competition
from a variety of businesses, including large fully integrated
biopharmaceutical and pharmaceutical companies and others that may
have greater financial, technical and human resources.
|
|
c. |
A competitor may enter with a generic of an approved innovator
product.
|
|
• |
We may not be successful in
using and expanding our Morphomer and SupraAntigen proprietary
technology platforms.
|
|
• |
We operate in highly
competitive and rapidly changing industries, which may result in
others discovering, developing or commercializing competing
products before or more successfully than we do.
|
|
• |
Our future growth and ability
to compete depends on retaining our key personnel and recruiting
additional qualified personnel including members of our Executive
Committee.
|
Risks
related to our relationships with third parties:
|
• |
If we fail to maintain, or
realize the benefits from, our current strategic relationships with
our current and potential future license and collaboration partners
our financial condition may be materially adversely affected.
|
|
• |
We may seek to form additional
strategic alliances in the future with respect to our product
candidates, and if we do not realize the benefits of such
alliances, our business, financial condition, commercialization
prospects and results of operations may be materially adversely
affected.
|
|
• |
Our collaboration agreements
may make us an attractive acquisition target under certain
circumstances.
|
Risks
related to intellectual property:
|
• |
We or our licensing or
collaboration partners may not have sufficient patent terms to
protect our products and business effectively which may adversely
affect our product sales and technology development.
|
|
• |
If we fail to comply with the
obligations to obtain and maintain patent protection such as
compliance with intellectual property agreements, including those
under which we license intellectual property and other rights to or
from third parties, or otherwise experience disruptions to our
business relationships with our licensees, our licensors and
partners, we could lose intellectual property rights that are
important to our business.
|
|
• |
We may be subject to claims
challenging the inventorship of our patents and other intellectual
property.
|
Risks
related to our financial condition and capital requirements:
|
• |
We are a clinical stage
biopharmaceutical company with a history of losses. We anticipate
incurring losses for the foreseeable future. As such, if we fail to
obtain additional funding via product revenues, license and
collaboration agreements, equity offerings or other forms of
financing, we may need to delay, reduce or eliminate certain of our
product development programs.
|
Risks
related to the regulatory environment:
|
• |
We cannot give any assurance
that any of our product candidates will receive regulatory
approval, which is necessary before they can be
commercialized.
|
|
• |
Even if we obtain regulatory
approvals in one jurisdiction, we may not be able to obtain
approval in other jurisdictions. Additionally, we will be subject
to ongoing obligations and review which may result in significant
additional expense.
|
|
• |
We have conducted and may in
the future conduct clinical studies for our product candidates
outside the US, and the FDA and applicable foreign regulatory
authorities may not accept data from such studies.
|
|
• |
Enacted and future legislation
may increase the difficulty and cost for us to obtain marketing
approval of and commercialize our product candidates and may affect
the prices we may set.
|
Risks
related to our common shares:
|
• |
We have limited free float in
our common shares which may have a negative impact on the liquidity
and market price of our common shares.
|
|
• |
Certain of our existing
shareholders exercise significant control over us, and your or
other shareholders’ interests may conflict with the interests of
such shareholders.
|
|
• |
We are a Swiss corporation.
The rights of our shareholders may be different from the rights of
shareholders in companies governed by the laws of US
jurisdictions.
|
|
• |
We are a foreign private
issuer (FPI) and, as a result, we are not subject to US proxy rules
and are subject to Exchange Act reporting obligations that, to some
extent, are more lenient and less frequent than those of a US
domestic public company.
|
|
• |
As an FPI, and as permitted by
the listing requirements of Nasdaq, we rely on certain home country
governance practices rather than the corporate governance
requirements of Nasdaq. Should we lose our FPI status, we would be
required to comply with the Exchange Act’s domestic reporting
regime, which would cause us to incur significant legal, accounting
and other expenses.
|
|
• |
It is likely that we were a
PFIC for 2019 and 2020, and there can be no assurance that we were
not, or will not be, a PFIC for any other taxable year. If a US
investor held our common shares during any taxable year in which we
are or were a PFIC, the investor generally will be subject to
adverse US federal income tax consequences.
|
Risks related to our business
We
depend heavily on the success of our clinical and, to a lesser
extent, preclinical products. Our clinical product candidates
include ACI-35, ACI-24 for AD and DS, ACI-7104, semorinemab,
crenezumab, Morphomer Tau, PI-2620 and our a-syn PET tracer. If our
clinical studies are unsuccessful, if we or our collaboration
partners do not obtain regulatory approval or if we or our
collaboration partners are unable to commercialize ACI-35, ACI-24
for AD and DS, ACI-7104, semorinemab, crenezumab, Morphomer Tau,
PI-2620 and our a-syn PET tracer, or if we experience significant
delays in doing so, our business, financial condition and results
of operations will be materially adversely affected.
We currently have no products
approved for sale and have invested a significant portion of our
efforts and financial resources in the development of ACI-35,
ACI-24 for AD and DS, ACI-7104, semorinemab, crenezumab, Morphomer
Tau, PI-2620 and our a-syn PET tracer, all of which are in clinical
development as well as other preclinical candidates such as
Morphomer a-syn and inflammasome. Our ability to generate product
revenues, which we do not expect will occur for at least the next
several years, if ever, will depend heavily on successful clinical
development, obtaining regulatory approval and eventual
commercialization of these product candidates. In this regard, we
rely heavily on our collaboration partners for clinical development
of certain of our product candidates, and they may choose to
discontinue the clinical development process in certain cases. In
addition, we currently generate no revenues from sales of any drugs
or diagnostics, and we may never be able to develop or
commercialize a marketable drug or diagnostic. The success of our
current and future product candidates will depend on several
factors, including the following:
|
• |
completing preclinical and clinical studies that demonstrate
the efficacy, safety and clinical utility of our preclinical and
clinical product candidates;
|
|
• |
receiving marketing approvals from applicable regulatory
authorities;
|
|
• |
establishing commercial manufacturing capabilities;
|
|
• |
launching commercial sales, marketing and distribution
operations;
|
|
• |
acceptance of our product candidates by patients, the medical
community and third-party payors;
|
|
• |
a continued acceptable safety profile following
approval;
|
|
• |
competing effectively with other therapies or diagnostic
approaches; and
|
|
• |
obtaining, maintaining, enforcing and defending our
intellectual property rights and claims and not infringing on third
parties’ intellectual property rights.
|
If we or our collaboration partners
do not achieve one or more of these factors in a timely manner or
at all, we could experience significant delays or an inability to
successfully commercialize our current or future product
candidates, which would materially adversely affect our business,
financial condition and results of operations.
Results of
early clinical studies may not be predictive of future study
results.
Positive or
timely results from preclinical or early-stage clinical studies do
not ensure positive or timely results in mid-to late-stage clinical
studies or product approval by the US FDA, the European Medicines
Agency (EMA), or comparable foreign regulatory authorities.
Products that show positive preclinical or early clinical results
may not show sufficient safety or efficacy in later-stage clinical
studies and therefore may fail to obtain regulatory approvals. In
addition, preclinical and clinical data are often susceptible to
varying interpretations and analyses. Many companies that believed
their product candidates performed satisfactorily in preclinical
and clinical studies have nonetheless failed to obtain marketing
approval for the product candidates. The FDA, the EMA and
comparable foreign regulatory authorities have substantial
discretion in the approval process and in determining when or
whether regulatory approval will be obtained for any of our product
candidates. Even if we believe that the data collected from
clinical studies of our product candidates are promising, such data
may not be sufficient to support approval by the FDA, the EMA or
any other regulatory authority.
In some instances, there can be
significant variability in safety and/or efficacy results between
different studies of the same product candidate due to numerous
factors, including changes in study procedures set forth in
protocols, differences in the size and type of the patient
populations, adherence to the dosing regimen and other study
protocols, and the rate of dropout among clinical study
participants. In the case of our later-stage clinical product
candidates, results may differ in general on the basis of the
larger number of clinical study sites and the additional countries
and languages involved in these clinical studies.
Clinical studies may include
subject-reported outcomes, some of which may be captured with
electronic diaries. We have no assurance and cannot rely on past
experience that the high frequency of questioning is not
influencing the measured outcome. In addition, low compliance with
daily reporting requirements may impact the studies’ validity or
statistical power. We cannot assure you that any Phase 2, Phase 3
or other clinical studies that either we or our collaboration
partners may conduct will demonstrate consistent or adequate
efficacy and safety to obtain regulatory approval to market our
product candidates.
If we or our collaboration partners
are required to conduct additional clinical studies or other
testing of any of our current or future product candidates that we
or our collaboration partners develop, beyond the studies and
testing that we or our collaboration partners contemplate, if we or
our collaboration partners are unable to successfully complete
clinical studies of our product candidates or other testing, if the
results of these studies or tests are unfavorable or are only
modestly favorable, or if there are safety concerns associated with
our current or future product candidates, we may:
|
• |
be delayed in obtaining marketing approval for our product
candidates;
|
|
• |
not obtain marketing approval;
|
|
• |
obtain approval for indications or patient populations that
are not as broad as intended or desired;
|
|
• |
obtain approval with labeling that includes significant use or
distribution restrictions or significant safety warnings, including
boxed warnings;
|
|
• |
be subject to conditional approval or otherwise to additional
post-marketing studies or other requirements; or
|
|
• |
remove the product from the market after obtaining marketing
approval.
|
Our product development costs will
also increase if we experience delays in testing or receiving
marketing approvals and we may be required to obtain additional
funds to complete clinical studies. We cannot assure you that our
clinical studies will begin as planned or be completed on schedule,
if at all, or that we will not need to amend our studies after they
have begun. Significant clinical study delays could also shorten
any periods during which we or our collaboration partners may have
the exclusive right to commercialize our product candidates, or
allow our competitors to bring products to market before we do,
which may harm our business and results of operations. In addition,
some of the factors that cause, or lead to, clinical study delays
may ultimately lead to the denial of regulatory approval of our
product candidates.
We may undertake one or more significant corporate transactions
that may not achieve their intended results, may adversely affect
our financial condition and our results of operations or result in
unforeseeable risks to our business.
We
continuously evaluate the acquisition or disposition of operating
businesses and assets and may in the future undertake one or more
significant transactions, such as our purchase of Affiris AG’s
(Affiris) program portfolio of therapeutics targeting a-syn,
notably PD01, a clinically-validated active vaccine candidate for
the treatment of Parkinson’s disease (PD). Any such transaction
could be material to our business and could take any number of
forms, including mergers, joint ventures and the purchase of equity
interests. The consideration for such acquisitive transactions may
include, among other things, cash, common shares or equity
interests in us or our subsidiaries, or a contribution of equipment
to obtain equity interests, and in conjunction with a transaction
we might incur additional indebtedness. We also routinely evaluate
the benefits of disposing of certain of our assets.
These
transactions may present significant risks such as insufficient
revenue to offset liabilities assumed, potential loss of
significant revenue and income streams, increased or unexpected
expenses, inadequate return of capital, regulatory or compliance
issues, the triggering of certain covenants in our debt agreements
(including accelerated repayment) and unidentified issues not
discovered in due diligence. In addition, such transactions could
distract management from current operations. As a result of the
risks inherent in such transactions, we cannot guarantee that any
such transaction will ultimately result in the realization of its
anticipated benefits or that it will not have a material adverse
effect on our business, financial condition and results of
operations. If we were to complete such an acquisition,
disposition, investment or other strategic transaction, we may
require additional debt or equity financing that could result in a
significant increase in our amount of debt and our debt service
obligations or the number of outstanding common shares, thereby
diluting holders of our common shares outstanding prior to such
acquisition.
Additional competitors could enter the market with generic versions
of our products, which may result in a material decline in sales of
affected products.
Under the Drug Price Competition
and Patent Term Restoration Act of 1984, or the Hatch-Waxman Act, a
pharmaceutical manufacturer may file an abbreviated new drug
application (ANDA), seeking approval of a generic copy of an
approved innovator product. Under the Hatch-Waxman Act, a
manufacturer may also submit a new drug application (NDA) under
Section 505(b)(2) that references the FDA’s prior approval of the
innovator product. A 505(b)(2) NDA product may be submitted for a
new or improved version of the original innovator product.
Hatch-Waxman also provides for certain periods of regulatory
exclusivity, which preclude FDA approval (or in some circumstances,
FDA filing and reviewing) of an ANDA or 505(b)(2) NDA. These
include, subject to certain exceptions, the period during which an
FDA-approved drug is subject to orphan-drug exclusivity. In
addition to the benefits of regulatory exclusivity, an innovator
NDA holder may have patents claiming the active ingredient, product
formulation or an approved use of the drug, which would be listed
with the product in the FDA publication, “Approved Drug Products
with Therapeutic Equivalence Evaluations,” known as the “Orange
Book.” If there are patents listed in the Orange Book, a generic or
505(b)(2) applicant that seeks to market its product before
expiration of the patents must include in the ANDA what is known as
a “Paragraph IV certification,” challenging the validity or
enforceability of, or claiming non-infringement of, the listed
patent or patents. Notice of the certification must also be given
to the innovator, and if within 45 days of receiving notice the
innovator, in order to protect its patents, sues the company that
manufactures the generic, approval of the ANDA is stayed for 30
months, or as lengthened or shortened by the court.
Accordingly, if ACI-35, ACI-24 for
AD and DS, ACI-7104, semorinemab, crenezumab, Morphomer Tau,
PI-2620 and our a-syn PET tracer are approved,
competitors could file ANDAs for generic versions of ACI-35, ACI-24
for AD and DS, ACI-7104, semorinemab, crenezumab, Morphomer Tau,
PI-2620 and our a-syn PET tracer or 505(b)(2) NDAs that reference
ACI-35, ACI-24 for AD and DS, ACI-7104, semorinemab, crenezumab,
Morphomer Tau, PI-2620 and our a-syn PET tracer, respectively. If
there are patents listed in the Orange Book for ACI-35, ACI-24 for
AD and DS, ACI-7104, semorinemab, crenezumab, Morphomer Tau,
PI-2620 and our a-syn PET tracer, respectively, those ANDAs and
505(b)(2) NDAs would be required to include a certification for
each listed patent, indicating whether the ANDA applicant does or
does not intend to challenge the patent. We cannot predict whether
any patents issuing from our pending patent applications will be
eligible for listing in the Orange Book, how any generic competitor
would address such patents, whether we would sue on any such
patents or the outcome of any such suit.
We may not be successful in
securing or maintaining proprietary patent protection for products
and technologies we develop or license. Moreover, if any patents
that are granted and listed in the Orange Book are successfully
challenged by way of a Paragraph IV certification and subsequent
litigation, the affected product could immediately face generic
competition and its sales would likely decline rapidly and
materially. Should sales decline, we may have to write off a
portion or all of the intangible assets associated with the
affected product, and our results of operations and cash flows
could be materially and adversely affected.
The
successful commercialization of our product candidates will depend
in part on the extent to which governmental authorities and health
insurers establish adequate coverage and reimbursement levels and
pricing policies.
The successful commercialization of
our product candidates will depend, in part, on the extent to which
coverage and reimbursement for our products will be available from
government and health administration authorities, private health
insurers and other third-party payors. To manage healthcare costs,
many governments and third-party payors increasingly scrutinize the
pricing of new technologies and require greater levels of evidence
of favorable clinical outcomes and cost-effectiveness before
extending coverage. In light of such challenges to prices and the
requirement for increasing levels of evidence of the benefits and
clinical outcomes of new technologies, we cannot be sure that
coverage will be available for any of our current or future product
candidates that we or our collaboration partners will commercialize
or, if available, that the reimbursement rates will be adequate in
each respective region. If we are unable to obtain adequate levels
of coverage and reimbursement for our product candidates, their
marketability will be negatively and materially impacted.
Third-party payors may deny
coverage and reimbursement status altogether for a given drug
product, or may cover the product but also establish prices at
levels that are too low to enable us to realize an appropriate
return on our investment in product development. Because the rules
and regulations regarding coverage and reimbursement change
frequently, in some cases at short notice, even when there is
favorable coverage and reimbursement, future changes may occur that
adversely impact the favorable status. Further, the net
reimbursement for drug products may be subject to additional
reductions in the future depending on policy changes enacted by the
US Congress.
The unavailability or inadequacy
and variability of third-party coverage and reimbursement could
have a material adverse effect on the market acceptance of our
product candidates and the future revenues we may expect to receive
from those products. In addition, we are unable to predict what
additional legislation or regulation relating to the healthcare
industry or third-party coverage and reimbursement may be enacted
in the future, or what effect such legislation or regulation would
have on our business.
Our
products may not gain market acceptance, in which case we or our
collaboration partners may not be able to generate product
revenues, which will materially adversely affect our business,
financial condition and results of operations.
Even if the FDA, the EMA or any
other regulatory authority approves the marketing of any product
candidates that we develop, physicians, healthcare providers,
patients or the medical community may not accept or use them.
Efforts to educate the medical community and third-party payors on
the benefits of our product candidates may require significant
resources and may not be successful. If any of our current or
future product candidates does not achieve an adequate level of
acceptance, we or our collaboration partners may not generate
significant product or royalty revenues or any profits from
operations. The degree of market acceptance of our product
candidates that are approved for commercial sale will depend on a
variety of factors, including:
|
• |
how clinicians and potential patients perceive our novel
products;
|
|
• |
the timing of market introduction;
|
|
• |
the number and clinical profile of competing products;
|
|
• |
our ability to provide acceptable evidence of safety and
efficacy or clinical utility;
|
|
• |
the prevalence and severity of any side effects;
|
|
• |
relative convenience and ease of administration;
|
|
• |
patient diagnostics and screening infrastructure in each
market;
|
|
• |
marketing and distribution support;
|
|
• |
availability of coverage, reimbursement and adequate payment
from health maintenance organizations and other third-party payors,
both public and private; and
|
|
• |
other potential advantages over alternative treatment
methods.
|
If our product candidates fail to
gain market acceptance, this will have a material adverse impact on
our ability to generate revenues to provide a satisfactory, or any,
return on our investments. Even if some products achieve market
acceptance, the market may prove to not be large enough to allow us
to generate significant revenues.
In addition, the potential market
opportunity of our product candidates is difficult to estimate
precisely. Our estimates of the potential market opportunity are
predicated on several key assumptions such as industry knowledge
and publications, third-party research reports and other surveys.
These assumptions involve the exercise of significant judgment on
the part of our management and are inherently uncertain, and the
reasonableness of these assumptions could not have been assessed by
an independent source in every detail. If any of the assumptions
proves to be inaccurate, then the actual market for our product
candidates could be smaller than our estimates of the potential
market opportunity. If the actual market for our product candidates
is smaller than we expect, or if any approved products fail to
achieve an adequate level of acceptance by physicians, healthcare
payors and patients, our product or royalty revenue may be limited
and it may be more difficult for us to achieve or maintain
profitability.
We
depend on enrollment of patients in our clinical studies for our
product candidates. If we are unable to enroll patients in our
clinical studies, our research and development efforts could be
materially adversely affected.
Successful and timely completion of
clinical studies will require that we enroll a sufficient number of
patient candidates. Studies may be subject to delays as a result of
patient enrollment taking longer than anticipated or by patient
withdrawal. Patient enrollment depends on many factors, including
the size and nature of the patient population, the eligibility
criteria for the study, the proximity of patients to clinical
sites, the design of the clinical protocol, the existence of
competing clinical studies, the availability of new drugs approved
for the indication the clinical study is investigating, and
clinicians’ and patients’ perceptions as to the potential
advantages of the drug being studied in relation to other available
therapies.
Generally, the specific target
population of patients and therapeutic time windows may make it
difficult for us to enroll enough patients to complete clinical
studies for our product candidates in a timely and cost-effective
manner. Delays in the completion of any clinical study of our
product candidates will increase our costs, slow down our product
candidate development and approval process, and delay or
potentially jeopardize our or our collaboration partners’ ability
to commence product sales and generate revenue. In addition, many
of the factors that cause, or lead to, a delay in the commencement
or completion of clinical studies may also ultimately lead to the
denial of regulatory approval of our product candidates.
If
serious adverse, undesirable or unacceptable side effects are
identified during the development of our product candidates or
following approval, if any, we may need to abandon our development
of such product candidates, the commercial profile of any approved
label may be limited, or we may be subject to other significant
negative consequences following marketing approval, if any.
If our product candidates are
associated with serious adverse, undesirable or unacceptable side
effects, we may need to abandon their development or limit
development to certain uses or subpopulations in which such side
effects are less prevalent, less severe or more acceptable from a
risk–benefit perspective. Many compounds that initially showed
promise in preclinical or early-stage testing were later found to
cause side effects that restricted their use and prevented further
development of the compound for larger indications.
Occurrence of serious procedure- or
treatment-related side effects could impede clinical study
enrollment and receipt of marketing approval from the FDA, the EMA
and comparable foreign regulatory authorities. Adverse events (AEs)
could also adversely affect physician or patient acceptance of our
product candidates.
Additionally, if one or more of our
product candidates receives marketing approval, and we or others
later identify undesirable side effects caused by such products, a
number of potentially significant negative consequences could
result, including the following:
|
• |
regulatory authorities may withdraw approvals of such product
and require us or our collaboration partners to take any approved
products off the market;
|
|
• |
regulatory authorities may require the addition of labeling
statements, specific warnings, a contraindication or field alerts
to physicians and pharmacies;
|
|
• |
we may be required to create a medication guide outlining the
risks of such side effects for distribution to patients;
|
|
• |
we may be required to change the way the product is
administered, to conduct additional studies or to change the
labeling of the product;
|
|
• |
we or our collaboration partners may be subject to limitations
in how we promote the product;
|
|
• |
sales of the product may decrease significantly;
|
|
• |
we could be sued and held liable for harm caused to patients;
and
|
|
• |
our reputation and physician or patient acceptance of our
products may suffer.
|
Any of these events could prevent us from achieving or
maintaining market acceptance of the particular product candidate,
if approved, and could significantly harm our business, results of
operations and prospects.
We
operate in highly competitive and rapidly changing industries,
which may result in others discovering, developing or
commercializing competing products before or more successfully than
we do.
The biopharmaceutical and
pharmaceutical industries are highly competitive and subject to
significant and rapid technological change. Our success is highly
dependent on our ability to discover, develop and obtain marketing
approval for new and innovative products on a cost-effective basis
and to market them successfully. In doing so, we face and will
continue to face intense competition from a variety of businesses,
including large, fully integrated pharmaceutical companies,
specialty pharmaceutical companies and biopharmaceutical companies,
academic institutions, government agencies and other private and
public research institutions in Europe, the US and other
jurisdictions. Many of our potential competitors, alone or with
their strategic partners, have substantially greater financial,
technical and human resources than we do and significantly greater
experience in the discovery and development of product candidates,
obtaining FDA and other regulatory approvals of treatments, and the
commercialization of those treatments. Mergers and acquisitions in
the pharmaceutical and biopharmaceutical industries may result in
even more resources being concentrated among a smaller number of
our competitors.
The highly competitive nature of
and rapid technological changes in the pharmaceutical and
biopharmaceutical industries could render our product candidates or
our technology obsolete or noncompetitive. The commercial
opportunity for our products could be reduced or eliminated if our
competitors:
|
• |
develop and commercialize products that are safer, more
effective, less expensive, or more convenient or easier to
administer;
|
|
• |
obtain quicker FDA or other regulatory approval for their
products;
|
|
• |
establish superior intellectual property and proprietary
positions;
|
|
• |
have access to more manufacturing capacity;
|
|
• |
implement more effective approaches to sales, marketing and
distribution; or
|
|
• |
form more advantageous strategic alliances.
|
Should any of these occur, our
business, financial condition and results of operations could be
materially adversely affected.
We believe that our key competitor
product candidates are (i) AADvac1 (Axon Neuroscience) for
ACI-35.030; (ii) UB-311 (Vaxxinity) and ABvac40 (Araclon Biotech)
for ACI-24; (iii) UB-312 (Vaxxinity) for ACI-7104; (iv) bepranemab
(UCB/Roche) and JNJ-63733657 (Janssen) for semorinemab; (v) ADUHELM
(Biogen), gantenerumab (Roche), lecanemab (BioArctic/Eisai),
donanemab (Eli Lilly and Company) and solanezumab (Eli Lilly and
Company) for crenezumab; (vi) TRx0237 (TauRx Pharmaceuticals) for
Morphomer Tau; and (vii) Tauvid (Eli Lilly and Company), APN-1607
(Aprinoia Therapeutics), MK-6240 (Cerveau/Merck) and GTP1
(Genentech) for PI-2620, as described under “Item 4. Information on
the Company—B. Business overview—Competition.”
We may
not be successful in our efforts to use and expand our Morphomer
and SupraAntigen proprietary technology platforms to build
additional product candidates for our pipeline.
A key element of our strategy is to
use and expand our Morphomer and SupraAntigen proprietary
technology platforms to create unique therapies and diagnostics for
conformational diseases, such as AD, and progress these product
candidates through clinical development. Although our research and
development efforts to date have resulted in a pipeline of product
candidates, we may not be able in the future to develop product
candidates that are safe and effective. Even if we are successful
in continuing to build our pipelines, the potential product
candidates that we identify may not be suitable for clinical
development, potentially as a result of having harmful side effects
or other characteristics indicating they may be unlikely to receive
marketing approval and achieve market acceptance.
Our
business is subject to economic, political, regulatory and other
risks associated with international operations.
Our business is subject to risks
associated with conducting business internationally. We and a
number of our suppliers and collaborative and clinical study
relationships are located outside the US. Accordingly, our future
results could be harmed by a variety of factors, including:
|
• |
economic weakness, including inflation, or political
instability in particular non-US economies and markets;
|
|
• |
differing regulatory requirements for drug approvals in non-US
countries;
|
|
• |
potentially reduced protection for intellectual property
rights;
|
|
• |
difficulties in compliance with non-US laws and
regulations;
|
|
• |
changes in non-US regulations and customs, tariffs and trade
barriers;
|
|
• |
changes in non-US currency exchange rates and currency
controls;
|
|
• |
changes in a specific country’s or region’s political or
economic environment;
|
|
• |
trade protection measures, import or export licensing
requirements or other restrictive actions such as sanctions by US
or non-US governments;
|
|
• |
negative consequences from changes in tax laws;
|
|
• |
compliance with tax, employment, immigration and labor laws
for employees living or traveling abroad;
|
|
• |
workforce uncertainty in countries where labor unrest is more
common than in the US;
|
|
• |
difficulties associated with staffing and managing
international operations, including differing labor
relations;
|
|
• |
production shortages resulting from any events affecting raw
material supply or manufacturing capabilities abroad; and
|
|
• |
business interruptions resulting from geopolitical actions,
including war and terrorism, or natural disasters including
earthquakes, typhoons, floods and fires.
|
Moreover, at the end of 2021 and
into 2022, tensions between the United States and Russia escalated
when Russia amassed large numbers of military ground forces and
support personnel on the Ukraine-Russia border and, in February
2022, Russia invaded Ukraine. In response, NATO has deployed
additional military forces to Eastern Europe, including to
Lithuania, and the Biden administration announced certain sanctions
against Russia. The invasion of Ukraine and the retaliatory
measures that have been taken, or could be taken in the future, by
the United States, NATO, and other countries have created global
security concerns that could result in a regional conflict and
otherwise have a lasting impact on regional and global economies,
any or all of which could disrupt our supply chain, adversely
affect our ability to conduct ongoing and future clinical trials of
our product candidates, and adversely affect our ability to
commercialize our products (subject to regulatory approval) in this
region. Currently, none of our clinical development or business
activities are conducted directly or otherwise in Russia or
Ukraine.
The Covid-19 pandemic has adversely impacted, and may continue to
impact, our business, including preclinical and clinical trials and
regulatory approvals.
In December
2019, a novel strain of coronavirus, Covid-19, surfaced in Wuhan,
Hubei Province, China. By March 2020, Covid-19 had spread to other
countries, including Switzerland and the United States, and was
declared a pandemic by the World Health Organization on March 11,
2020. Since the beginning of the pandemic, governments, public
institutions, and other organizations in countries and localities
where Covid-19 cases have been identified are taking certain
preventative or protective measures to combat the transmission of
the virus, including implementation of travel restrictions or bans,
closures of non-essential businesses, limitations of public
gatherings, other social distancing and shelter-in-place measures,
and delays or cancellations of elective surgeries. The Covid-19
pandemic poses the risk that the Company, our employees,
contractors, suppliers, and other partners may be prevented from
conducting business activities for an indefinite period of time due
to shutdowns that may be requested or mandated by state and federal
governmental authorities.
As Covid-19
continues to spread around the globe, we have experienced
disruptions impacting our business and clinical trials and we may
continue to experience disruptions that could materially impact our
business and planned clinical trials, including:
|
• |
delays or difficulties in
conducting preclinical research and clinical trials;
|
|
• |
interruption in global manufacturing and shipping that may
affect the manufacturing and/or transport of clinical trial
materials and other materials, including testing equipment and
personal protective equipment, used at our or our contract research
organizations’ (CROs’) and contract manufacturing organizations’
(CMOs’) facilities;
|
|
• |
changes in local regulations as part of a response to the
Covid-19 coronavirus outbreak which may require us to change the
way in which clinical trials are conducted and may result in
unexpected costs; and
|
|
• |
impact our ability to secure
additional financing.
|
In addition, the outbreak of Covid-19 could
disrupt our operations due to absenteeism by infected or ill
members of Executive Management or other employees, or absenteeism
by members of Executive Management and other employees who elect
not to come to work due to the illness affecting others in our
office or laboratory facilities, or due to quarantines. Covid-19
illness could also impact members of our Board and its ability to
hold meetings. Further information concerning details of the
impact of Covid-19 on our programs can be found under “Item 5:
Operating and financial review and prospects.”
Our
ability to effectively monitor and respond to the rapid and ongoing
developments and expectations relating to environmental, social and
governance (ESG) matters, including related social expectations and
concerns, may impose unexpected costs on us or result in
reputational or other harm to us that could have a material adverse
effect on our business, financial condition and results of
operations.
If we are not
able to adequately recognize and respond to the rapid and ongoing
developments and governmental and social expectations relating to
ESG matters such as climate change and access to health care and
affordable drugs, this failure could result in missed corporate
opportunities, additional regulatory, social or other scrutiny of
us and our business, the imposition of unexpected costs or in
damage to our reputation or our various relationships with
governments, customers, employees, third parties and the
communities in which we operate, in each case that could have a
material adverse effect on our business, financial condition and
results of operations.
We have
no history of commercializing biologics or pharmaceutical products,
which may make it difficult to evaluate the prospects for our
future viability.
We began our operations in 2003.
Our operations to date have been limited to financing and staffing
our company, developing our technology and developing our product
candidates as well as early-stage clinical trials. We have not yet
demonstrated an ability to successfully complete a large-scale,
pivotal clinical study, obtain marketing approval, manufacture a
commercial-scale product, or conduct sales and marketing activities
necessary for successful product commercialization. Consequently,
predictions about our future success or viability may not be as
accurate as they could be if we had a history of successfully
developing and commercializing biologics or pharmaceutical
products.
Our
future growth and ability to compete depends on retaining our key
personnel and recruiting additional qualified personnel.
Our success depends upon the
continued contributions of our key management, scientific and
technical personnel, many of whom have substantial experience with
or been instrumental for us and our projects. Members of our key
management include Dr. Andrea Pfeifer, our Chief Executive Officer;
Dr. Marie Kosco-Vilbois, our Chief Scientific Officer; Dr. Johannes
Rolf Streffer, our Chief Medical Officer; Piergiorgio Donati, our
Chief Technical Operations Officer; Joerg Hornstein, our Chief
Financial Officer; Jean-Fabien Monin, our Chief Administrative
Officer; Dr. Julien Rongère, our Vice President (VP) for Regulatory
Affairs and Quality Assurance; Dr. Olivier Sol, our VP Head of
Clinical Development; Dr. Bojana Portmann, our Associate Vice
President for Intellectual Property and Business Development (AVP
IP); Alexandre Caratsch, our General Counsel; and Mark Danton, our
VP Information Systems, Security and Digital Technologies.
The loss of our key managers and
senior scientists could delay our research and development
activities. Laws and regulations on executive compensation,
including legislation in our home country, Switzerland, may
restrict our ability to attract, motivate and retain the required
level of qualified personnel. In Switzerland, legislation affecting
public companies is in force that, among other things, imposes an
annual binding shareholders’ “say on pay” vote with respect to the
total compensation of executive management, including executive
officers and the board of directors, and prohibits severance or
similar payment, bonuses for company purchases and sales, and
additional contracts as consultants to or employees of other
companies in the group. In addition, the competition for qualified
personnel in the pharmaceutical and biopharmaceutical field is
intense, and our future success depends upon our ability to
attract, retain and motivate highly skilled scientific, technical
and managerial employees. We face competition for personnel from
other companies, universities, public and private research
institutions and other organizations. If our recruitment and
retention efforts are unsuccessful in the future, it may be
difficult for us to implement our business strategy, which could
have a material adverse effect on our business.
We may
become exposed to costly and damaging liability claims, either when
testing our product candidates in the clinic or at the commercial
stage or as a result of claims against our directors and officers;
and our liability insurance may not cover all damages from such
claims.
We are exposed to potential product
liability and professional indemnity risks that are inherent in the
research, development, manufacturing, marketing and use of
pharmaceutical or biopharmaceutical products. Currently we have no
products that have been approved for commercial sale; however, our
current and future use of product candidates in clinical studies,
and the sale of any approved products in the future, may expose us
to liability claims. These claims might be made by patients that
use the product, by healthcare providers, or by pharmaceutical or
biopharmaceutical companies or others selling such products. Any
claims against us, regardless of their merit, could be difficult
and costly to defend and could materially adversely affect the
market for our product candidates or any prospects for
commercialization of our product candidates.
Although the clinical study process
is designed to identify and assess potential side effects, it is
always possible that a drug, even after regulatory approval, may
exhibit unforeseen side effects. If any of our product candidates
were to cause adverse side effects during clinical studies or after
approval of the product candidate, we may be exposed to substantial
liabilities. Physicians and patients may not comply with any
warnings that identify known potential adverse effects and patients
who should not use our product candidates.
We purchase liability insurance in
connection with the clinical studies that we undertake and for
purposes of indemnifying our directors and officers for claims
against them in amounts that we consider to be consistent with
industry norms. It is possible that our liabilities could exceed
our insurance coverage. For example, we intend to expand our
insurance coverage to include the sale of commercial products if we
obtain marketing approval for any of our product candidates.
However, we may not be able to maintain insurance coverage at a
reasonable cost or obtain insurance coverage that will be adequate
to satisfy any liability that may arise. If a successful liability
claim or series of claims is brought against us for uninsured
liabilities or in excess of insured liabilities, our assets may not
be sufficient to cover such claims and our business operations
could be impaired.
Should any of the events described
above occur, this could have a material adverse effect on our
business, financial condition and results of operations.
We may
seek to obtain orphan-drug designation for certain of our product
candidates. Orphan-drug designation may not ensure that we will
enjoy market exclusivity in a particular market, and if we fail to
obtain or maintain orphan-drug exclusivity for such product
candidates, we may be subject to earlier competition and our
potential revenue will be reduced.
Under the Orphan Drug Act, the FDA
may designate a product as an orphan drug if it is intended to
treat a rare disease or condition, defined as a patient population
of fewer than 200,000 in the US, or a patient population greater
than 200,000 in the US where there is no reasonable expectation
that the cost of developing the drug will be recovered from sales
in the US. In the European Union (EU), the EMA’s Committee for
Orphan Medicinal Products grants orphan-drug designation to promote
the development of products that meet the following criteria: a)
they are intended for the diagnosis, prevention, or treatment of a
life-threatening or chronically debilitating condition affecting
not more than 5 in 10,000 persons in the EU or for products that
are intended for the diagnosis, prevention or treatment of a
life-threatening, seriously debilitating or serious and chronic
condition when, without incentives, it is unlikely that sales of
the drug in the EU would be sufficient to justify the necessary
investment in developing the drug or biological product; and b)
there is no satisfactory method of diagnosis, prevention, or
treatment, or, if such a method exists, the medicine must be of
significant benefit to those affected by the condition.
In the US, orphan-drug designation
entitles a party to financial incentives such as opportunities for
grant funding toward clinical study costs, tax advantages and
user-fee waivers. In addition, if a product receives the first FDA
approval for the indication for which it has orphan designation,
the product is entitled to orphan-drug exclusivity, which means
that the FDA cannot approve any other application to market the
same drug for the same indication for a period of 7 years, except
in limited circumstances, such as a showing of clinical superiority
over the product with orphan exclusivity or if the manufacturer is
unable to assure sufficient product quantity. In the EU,
orphan-drug designation entitles a party to financial incentives
such as reduction of fees or fee waivers and 10 years of market
exclusivity for the orphan indication following drug or biological
product approval, provided that the criteria for orphan designation
are still applicable at the time of the granting of the marketing
authorization. This period may be reduced to 6 years if at the end
of the fifth year, the orphan-drug designation criteria are no
longer met, including where it is shown that the product is
sufficiently profitable not to justify maintenance of market
exclusivity.
We may not be able to obtain
orphan-drug designation for any of our product candidates, and even
if we do, we may not be the first to obtain marketing approval for
any particular orphan indication due to the uncertainties
associated with developing pharmaceutical or biopharmaceutical
products. Further, even if we obtain orphan-drug designation for a
product, that exclusivity may not effectively protect the product
from competition because different drugs with different active
moieties can be approved for the same condition. Orphan-drug
designation neither shortens the development time or regulatory
review time of a drug nor gives the drug any advantage in the
regulatory review or approval process.
Due to
our limited resources and access to capital, we must prioritize
development of certain product candidates.
Because we have limited resources
and access to capital to fund our operations, we must decide which
product candidates to pursue and the amount of resources to
allocate to each. Our decisions concerning the allocation of
research, collaboration, management and financial resources toward
particular compounds, product candidates or therapeutic areas may
not lead to the development of viable commercial products and may
divert resources away from better opportunities. Similarly, our
potential decisions to delay, terminate or collaborate with third
parties in respect of certain product development programs may also
prove not to be optimal and could cause us to miss valuable
opportunities. If we make incorrect determinations regarding the
market potential of our product candidates or misread trends in the
pharmaceutical or biopharmaceutical industry, in particular for
neurological disorders, our business, financial condition and
results of operations could be materially adversely affected.
Our
research and development activities could be affected or delayed as
a result of possible restrictions on animal testing.
Certain laws and regulations
require us to test our product candidates on animals before
initiating clinical studies in humans. Animal testing activities
have been the subject of controversy and adverse publicity. Animal
rights groups and other organizations and individuals have
attempted to stop animal testing activities by pressing for
legislation and regulation in these areas and by disrupting these
activities through protests and other means. To the extent that the
activities of these groups are successful, our research and
development activities may be interrupted, delayed or become more
expensive.
A breakdown or breach of our information technology systems and
cybersecurity efforts, or those of our key business partners, CROs
or service providers, could subject us to liability or reputational
damage or interrupt the operation of our business.
We are increasingly dependent upon
technology systems and data. Our computer systems continue to
increase in multitude and complexity due to the growth in our
business, making them potentially vulnerable to breakdown,
malicious intrusion and
random attack. Despite the implementation of security
measures, our internal computer systems and those of our key
business partners, CROs and service providers may be vulnerable to
damage from computer viruses, unauthorized access or other similar
cyber-attacks or incidents. If such an event were to occur and
cause interruptions in our operations, it could result in a
material disruption of our development programs and our business
operations.
Data privacy or security breaches,
cyber-attacks and other cybersecurity incidents, including those by
individuals authorized to access our technology systems or others,
may pose a risk that sensitive data, including intellectual
property, trade secrets or personal information belonging to us,
our patients, study subjects or other business partners, may be
exposed to unauthorized persons or to the public. Cyber-attacks are
increasing in their frequency, sophistication and intensity, and
are becoming increasingly difficult to detect. They are often
carried out by motivated, well-resourced, skilled and persistent
actors, including nation states, organized crime groups,
“hacktivists” and employees or contractors acting with malicious
intent. Cyber-attacks could include the deployment of harmful
malware and key loggers, ransomware, a denial-of-service attack, a
malicious website, phishing attacks, computer viruses, social
engineering and other means to affect the confidentiality,
integrity and availability of our technology systems and data. Our
key business partners, CROs and service providers face similar
risks and any security breach of their systems could adversely
affect our security posture. For example, the loss of
clinical trial data from completed, ongoing or future clinical
trials could result in delays in our regulatory approval efforts
and significantly increase our costs to recover or reproduce the
data. Likewise, we rely on our third-party research institution
collaborators for research and development of our product
candidates and on other third parties for the manufacture of our
product candidates and to conduct clinical trials, and similar
events relating to their computer systems could also have a
material adverse effect on our business. Our ability to monitor our
CROs’, contractors’ and consultants’ data security practices are
limited, and due to applicable laws and regulations or contractual
obligations, we may be held responsible for any security breaches
or cybersecurity attack attributed to them as they relate to the
information we share with them. To the extent that any disruption
or security breach were to result in a loss of, or damage to, our
data or systems, or inappropriate disclosure of confidential or
proprietary information or personal data of our employees, partners
or study subjects, we could incur liability including notification
obligations (including to the impacted individuals and applicable
regulators or supervisory authorities), and the further development
and commercialization of our product candidates could be
delayed.
Although we continue to build and improve
our systems and infrastructure, and to implement technical,
organizational and legal security measures, and believe we have
taken appropriate security measures to reduce these risks to our
data and information technology systems, there can be no assurance
that our efforts will prevent, detect or appropriately respond to
breakdowns or breaches in our systems that could adversely affect
our business and operations and/or result in the loss of critical
or sensitive information, including personal information, which
could result in financial, legal, business or reputational harm to
us. We continue to invest in industry standard IS/IT solutions and
managed services that often include the relevant, layered
protection and monitoring practices surrounding our data and IT
systems and related infrastructure. These investments reduce
further these risks in that they enable organizations such as ours
to leverage the resources necessary to monitor IT systems and
infrastructure for any current or potential threats. We also
regularly perform risk and impact assessments, the results of which
generally lead to the implementation of certain measures designed
to increase our level of data protection. These investments are
costly, and as cyber threats continue to evolve, we may be required
to expend significant, additional resources to continue to modify
and/or enhance our protective, detective and responsive measures
required to remediate any identified information security
vulnerabilities. In addition, our liability insurance may not be
sufficient in type or amount to cover us against claims related to
security breaches, cyber-attacks and other related breaches.
We may be required to expend significant capital and other
resources to protect against and respond to any attempted or
existing cybersecurity incidents. In addition, our remediation
efforts may not be successful.
Changes
in laws, rules or regulations relating to data privacy and
security, or any actual or perceived failure by us to comply with
such laws, rules, regulations and standards, or contractual or
other obligations relating to data privacy and security, could
result in claims, changes to our business practices, penalties,
increased cost of operations and could have a material adverse
effect on our reputation, results of operations, financial
condition and cash flows.
We are, and may increasingly
become, subject to various laws, rules, regulations and standards,
as well as contractual obligations, relating to data privacy and
security in the jurisdictions in which we operate. The regulatory
environment related to data privacy and security is increasingly
rigorous, with new and constantly changing requirements applicable
to our business, and enforcement practices are likely to remain
uncertain for the foreseeable future. These laws, rules,
regulations and standards may be interpreted and applied
differently over time and from jurisdiction to jurisdiction and in
a manner that is inconsistent with our data practices and that
could have a material adverse effect on our results of operations,
financial condition and cash flows. New laws, amendments to or
reinterpretations of existing laws, rules, regulations, standards
and other obligations may require us to incur additional costs and
restrict our business operations, and may require us to change how
we use, collect, store, transfer or otherwise process certain types
of personal information and to implement new processes to comply
with those laws.
In the US, there are numerous
federal and state laws and regulations related to the privacy and
security of personal information. Regulations promulgated pursuant
to the US Health Insurance Portability and Accountability Act of
1996 (HIPAA) establish privacy and security standards that limit
the use and disclosure of protected health information, and require
the implementation of administrative, physical and technological
safeguards to protect the privacy of protected health information
and to ensure the confidentiality, integrity and availability of
electronic protected health information. Determining whether
protected health information has been handled in compliance with
applicable privacy standards and our contractual obligations can be
complex and may be subject to changing interpretation. Numerous
states have enacted or are in the process of enacting state level
data privacy laws and regulations governing the collection, use,
and other processing of state residents’ personal information, such
as the California Consumer Privacy Act (CCPA), which took effect on
January 1, 2020 and provides new and enhanced data privacy rights
to California residents, such as affording California residents the
right to access and delete their information and to opt out of
certain sharing and sales of personal information, and the
California Privacy Rights Act of 2020 (CPRA), which is effective in
most material respects starting on January 1, 2023 and imposes
additional obligations on covered companies and will significantly
modify the CCPA. In addition, laws in all 50 states require
businesses to provide notice to individuals whose personal
information has been disclosed as a result of a data breach.
Internationally, laws, regulations
and standards in many jurisdictions apply broadly to the
collection, use, retention, security, disclosure, transfer and
other processing of personal information. For example, the EU
General Data Protection Regulation (GDPR), which became effective
in May 2018, greatly increased the European Commission’s
jurisdictional reach of its laws and adds a broad array of
requirements for handling personal data. EU Member States are
tasked under the GDPR to enact, and to have enacted, certain
implementing legislation that adds to and/or further interprets the
GDPR requirements and potentially extends our obligations and
potential liability for failing to meet such obligations. The GDPR,
together with national legislation, regulations and guidelines of
the EU Member States and Switzerland (via its Federal Data
Protection Act) governing the processing of personal data, impose
strict obligations and restrictions on the ability to collect, use,
retain, protect, disclose, transfer and otherwise process personal
data. In particular, the GDPR includes obligations and restrictions
concerning the consent and rights of individuals to whom the
personal data relates (and the obligations of sponsors of clinical
trials acting as data controllers), the transfer of personal data
out of the European Economic Area (EEA), the notification of
security breaches and the security and confidentiality of personal
data. The GDPR authorizes fines for certain violations of up to 4%
of global annual revenue or EUR 20 million, whichever is greater.
The GDPR also applies to our key business partners, CROs and
service providers, whether or not they are located in Europe, with
which we share personal data subject to the GDPR. Additionally,
following Brexit, we also are subject to the UK General Data
Protection Regulation (UK GDPR) (i.e., a version of the GDPR as
implemented into UK law), exposing us to two parallel regimes with
potentially divergent interpretations and enforcement actions for
certain violations. While the European Commission issued an
adequacy decision intended to last for at least four years in
respect of the UK’s data protection framework, enabling data
transfers from EU member states to the UK to continue without
requiring organizations to put in place contractual or other
measures in order to lawfully transfer personal data between the
territories, the relationship between the UK and the EU in relation
to certain aspects of data privacy and security law remains
unclear. Although we do not have material operations in the UK, we
cannot rule out potential disruptions in relation to the clinical
regulatory framework applicable to our clinical studies in the UK,
and to data privacy and security rules with respect to personal
data sharing with vendors and clinical investigators in the UK, and
we cannot predict future implications.
All of these evolving compliance
and operational requirements impose significant costs, which are
likely to increase over time. In addition, such requirements may
require us to modify our data processing practices and policies,
distract management or divert resources from other initiatives and
projects. For instance, the European Union Court of Justice and the
Swiss Data Protection Authority have declared the US Privacy Shield
to be inadequate for transfers of personal data out of the EU and
Switzerland, which could increase our compliance burden. If we are
unable to properly protect the privacy and security of personal
information, including protected health information, we could be
found to have breached our contracts. In addition, any failure or
perceived failure by us to comply with any applicable federal,
state or similar foreign laws and regulations relating to data
privacy and security could result in damage to our reputation and
our relationship with our customers, as well as proceedings or
litigation by governmental agencies, customers, partners,
collaborators and/or study subjects, including class action privacy
litigation in certain jurisdictions, which would subject us to
significant fines, sanctions, awards, penalties or judgments, all
of which could have a material adverse effect on our business,
results of operations, financial condition and prospects.
Business
disruptions could seriously harm our future revenue and financial
condition and increase our costs and expenses.
Our operations, and those of our
third-party research institution collaborators, CROs, CMOs,
suppliers, and other contractors and consultants, could be subject
to earthquakes, power shortages, telecommunications failures, water
shortages, floods, hurricanes, typhoons, fires, extreme weather
conditions, medical epidemics, and other natural or man-made
disasters or business interruptions, for which we are partly
uninsured. In addition, we rely on our third-party research
institution collaborators for conducting research and development
of our product candidates, and they may be affected by government
shutdowns or withdrawn funding. Moreover, at the end of 2021 and
into 2022, tensions between the United States and Russia escalated
when Russia amassed large numbers of military ground forces and
support personnel on the Ukraine-Russia border and, in February
2022, Russia invaded Ukraine. In response, North Atlantic Treaty
Organization, or NATO has deployed additional military forces to
Eastern Europe, including to Lithuania, and the Biden
administration announced certain sanctions against Russia. The
invasion of Ukraine and the retaliatory measures that have been
taken, or could be taken in the future, by the United States, NATO,
and other countries have created global security concerns that
could result in a regional conflict and otherwise have a lasting
impact on regional and global economies, any or all of which could
disrupt our supply chain, adversely affect our ability to conduct
ongoing and future clinical trials of our product candidates, and
adversely affect our ability to commercialize our products (subject
to regulatory approval) in this region. Currently, none of our
clinical development or business activities are conducted directly
or otherwise in Russia or Ukraine. The occurrence of any of these
business disruptions could seriously harm our operations and
financial condition and increase our costs and expenses. We rely on
third-party manufacturers to produce and process our product
candidates. Our ability to obtain clinical supplies of our product
candidates could be disrupted if the operations of these suppliers
are affected by a man-made or natural disaster or by other business
interruption.
The vast majority of our operations
including our corporate headquarters are located in Ecublens, near
Lausanne, Canton of Vaud, Switzerland. Damage or extended periods
of interruption to our corporate, development or research
facilities due to fire, natural disaster, power loss,
communications failure, unauthorized entry or other events could
cause us to cease or delay development of some or all of our
product candidates. Although we maintain property damage and
business interruption insurance coverage on these facilities, our
insurance might not cover all losses under such circumstances and
our business may be seriously harmed by such delays and
interruption.
We have
never commercialized a product candidate before and may lack the
necessary expertise, personnel and resources to successfully
commercialize our products on our own or together with suitable
partners.
We have never commercialized a
product candidate, and we currently have no sales force, marketing
or distribution capabilities. To achieve commercial success for our
product candidates, we will have to develop our own sales,
marketing and supply organization or outsource these activities to
third parties.
Factors that may affect our ability
to commercialize our product candidates on our own include
recruiting and retaining adequate numbers of effective sales and
marketing personnel, obtaining access to or persuading adequate
numbers of physicians to prescribe our product candidates, and
other unforeseen costs associated with creating an independent
sales and marketing organization. Developing a sales and marketing
organization requires significant investment, is time-consuming and
could delay the launch of our product candidates. We may not be
able to build an effective sales and marketing organization. In
addition, successful commercialization also requires an enhanced
regulatory organization which we currently do not have. If we are
unable to build our own distribution and marketing capabilities,
are unable to find suitable partners for the commercialization of
our product candidates or do not successfully obtain the necessary
regulatory capabilities, we may not generate revenues from them or
be able to reach or sustain profitability.
Risks related to our relationships
with third parties
If we
fail to maintain our current strategic relationships with
Genentech, a member of the Roche Group, Eli Lilly and Company
(Lilly), Janssen Pharmaceuticals Inc. (Janssen), Life Molecular
Imaging SA (LMI) (formerly Piramal Imaging SA) and other of our
current or future strategic partners, our business,
commercialization prospects and financial condition may be
materially adversely affected.
We have two partnerships with
Genentech. In 2006, we granted Genentech an exclusive, worldwide
license for crenezumab. In 2012, we entered into a second
partnership to commercialize anti-Tau antibodies for use as
immunotherapies. In December 2018, we signed a license agreement
with Lilly to research and develop Morphomer Tau small molecules
for the treatment of AD and other neurodegenerative diseases. This
collaboration commenced in Q1 2019. We are in a partnership with
Janssen to develop and commercialize therapeutic anti-Tau vaccines
for the treatment of AD and potentially other Tauopathies. We also
have a diagnostic partnership with LMI for one of our compounds
from our Morphomer chemical library, which bind pathological Tau
for use as a PET tracer. Our collaboration partners each have the
right to terminate their agreements with us for any reason upon
providing us with a certain notice period. If Genentech, Lilly,
Janssen, LMI or other of our current or future strategic partners
terminates its agreement with us at any time, it could delay or
prevent development of our product candidates and materially harm
our business, financial condition, commercialization prospects and
results of operations.
Good relationships with Genentech,
Lilly, Janssen, LMI and other of our current or future strategic
partners are important for our business prospects. If our
relationships with Genentech, Lilly, Janssen, LMI or other of our
current or future strategic partners were to deteriorate
substantially or if Genentech, Lilly, Janssen, LMI or other of our
current or future strategic partners were to challenge our use of
their intellectual property or our calculations of the payments we
are owed under our agreements, our business, financial condition,
commercialization prospects and results of operations could be
materially adversely affected.
Lastly, our collaboration
agreements with Genentech, Lilly, Janssen and LMI provide each
partner with control over, and responsibility for, the clinical
development process, including obtaining regulatory and marketing
approvals, manufacturing costs and sales and marketing costs. Our
other existing collaboration agreements provide our collaboration
partners with similar control over the clinical development
process, and future collaboration agreements may also relinquish
development control to our partners. Genentech or our other current
or future collaboration partners may and do separately pursue
competing products, therapeutic approaches or technologies to
develop treatments for the diseases targeted by us or our
collaborative efforts. Even if our partners continue their
contributions to the collaborative agreements to which we are a
party, they may nevertheless determine not to actively pursue the
development or commercialization of any resulting products. Our
partners may also fail to perform their obligations under the
collaboration agreements or may be slow in performing their
obligations. Any of these circumstances could result in a material
adverse impact on our business, financial condition,
commercialization prospects or results of operations.
We may
seek to form additional strategic alliances in the future with
respect to our product candidates, and if we do not realize the
benefits of such alliances, our business, financial condition,
commercialization prospects and results of operations may be
materially adversely affected.
Our product development programs
and the potential commercialization of our product candidates will
require substantial additional liquidity to fund expenses and may
require expertise, such as sales and marketing expertise, which we
do not currently possess. Therefore, in addition to our
relationships with Genentech, Lilly, Janssen and LMI, we may decide
to enter into strategic alliances or to create joint ventures or
collaborations with pharmaceutical or biopharmaceutical companies
for the further development and potential commercialization of
those and other of our product candidates.
We face significant competition in
seeking appropriate collaborators. Collaborations are complex and
time-consuming to negotiate, document and manage. Any delays in
entering into new strategic partnership agreements related to our
product candidates could delay the development and
commercialization of our product candidates and reduce their
competitiveness even if they reach the market. We may also be
restricted under existing and future collaboration agreements from
entering into strategic partnerships or collaboration agreements on
certain terms with other potential collaborators. We may not be
able to negotiate collaborations on acceptable terms, or at all,
for any of our existing or future product candidates and programs
because the potential partner may consider that our research and
development pipeline is insufficiently developed to justify a
collaborative effort, or that our product candidates and programs
do not have the requisite potential to demonstrate safety and
efficacy in the target population. If we are unsuccessful in
establishing and maintaining a collaboration with respect to a
particular product candidate, we may have to curtail the
development of that product candidate, reduce the scope of or delay
its development program or one or more of our other development
programs, delay its potential commercialization or reduce the scope
of our sales or marketing activities, or increase our expenditures
and undertake development or commercialization activities at our
own expense, for which we have not budgeted. If we elect to
increase our expenditures to fund development or commercialization
activities on our own, we may need to obtain additional capital,
which may not be available to us on acceptable terms or at all. If
we do not have sufficient funds, we will not be able to bring our
product candidates to market and generate product revenue. Even if
we are successful in establishing a new strategic partnership or
entering into a collaboration agreement, we cannot be certain that,
following such a strategic transaction or license, we will be able
to progress the development and commercialization of the applicable
product candidates as envisaged, or that we will achieve the
revenues that would justify such transaction, and we could be
subject to the following risks, each of which may materially harm
our business, commercialization prospects and financial
condition:
|
• |
we may not be able to control the amount and timing of
resources that the collaboration partner devotes to the product
development program;
|
|
• |
the collaboration partner may experience financial
difficulties;
|
|
• |
we may be required to grant or otherwise relinquish important
rights such as marketing, distribution and intellectual property
rights;
|
|
• |
a collaboration partner could move forward with a competing
product developed either independently or in collaboration with
third parties, including our competitors; or
|
|
• |
business combinations or significant changes in a
collaboration partner’s business strategy may adversely affect our
willingness to continue any arrangement.
|
We rely
on third parties to conduct our nonclinical and clinical studies
and perform other tasks for us. If these third parties do not
successfully carry out their contractual duties, meet expected
deadlines or comply with regulatory requirements, we may not be
able to obtain regulatory approval for or commercialize our product
candidates, and our business could be substantially harmed.
We have relied upon and plan to
continue to rely upon third-party clinical CROs, to monitor and
manage data for our ongoing nonclinical and clinical programs,
including the clinical studies of our product candidates. We rely
on these parties for execution of our nonclinical and clinical
studies and control only certain aspects of their activities.
Nevertheless, we are responsible for ensuring that each of our
studies is conducted in accordance with the applicable protocol,
legal, regulatory and scientific standards and our reliance on the
clinical CROs does not relieve us of our regulatory
responsibilities. We and our clinical CROs and other vendors are
required to comply with current Good Manufacturing Practice (cGMP),
current Good Clinical Practice (cGCP), and current Good Laboratory
Practice (cGLP), which are regulations and guidelines enforced by
the FDA, the Competent Authorities of the Member States of the EU
and comparable foreign regulatory authorities for our product
candidates in nonclinical and clinical development (where
applicable). Regulatory authorities enforce these regulations
through periodic inspections of study sponsors, principal
investigators, study sites and other contractors. If we or any of
our clinical CROs or vendors fail to comply with applicable
regulations, the data generated in our nonclinical and clinical
studies may be deemed unreliable and the EMA, FDA, other regulatory
authorities may require us to perform additional nonclinical and
clinical studies before approving our marketing applications. We
cannot assure you that upon inspection by a given regulatory
authority, such regulatory authority will determine that all of our
clinical studies comply with cGCP regulations. In addition, our
clinical studies must be conducted with products produced under
cGMP regulations. Our failure to comply with these regulations may
require us to repeat clinical studies, which would delay the
regulatory approval process.
If any of our relationships with
these third-party clinical CROs terminates, we may not be able to
enter into arrangements with alternative clinical CROs or do so on
commercially reasonable terms. In addition, our clinical CROs are
not our employees, and except for remedies available to us under
our agreements with such clinical CROs, we cannot control whether
or not they devote sufficient time and resources to our ongoing
nonclinical and clinical programs. If clinical CROs do not
successfully carry out their contractual duties or obligations or
meet expected deadlines, if they need to be replaced or if the
quality or accuracy of the data they obtain is compromised due to
their failure to adhere to our protocols, regulatory requirements,
or for other reasons, our clinical studies may be extended,
delayed, or terminated and we may not be able to obtain regulatory
approval for or successfully commercialize our product candidates.
Clinical CROs may also generate higher costs than anticipated. As a
result, our results of operations and the commercial prospects for
our product candidates would be harmed, our costs could increase,
and our ability to generate revenue could be delayed.
Switching or adding additional
clinical CROs involves additional cost and requires management time
and focus. In addition, there is a natural transition period when a
new clinical CRO commences work. As a result, delays occur, which
could materially impact our ability to meet our desired clinical
development timelines. Though we carefully manage our relationships
with our clinical CROs, there can be no assurance that we will not
encounter similar challenges or delays in the future or that these
delays or challenges will not have a material adverse impact on our
business, financial condition and prospects.
We
currently rely on third-party suppliers and other third parties for
production of our product candidates and our dependence on these
third parties may impair the advancement of our research and
development programs and the development of our product
candidates.
We currently rely on, and expect to
continue to rely on, third parties for the manufacturing and supply
of chemical and biological compounds and formulations for the
clinical studies of our current and future product candidates. For
the foreseeable future, we expect to continue to rely on such third
parties for the manufacture of any of our product candidates on a
clinical or commercial scale, if any of our product candidates
receives regulatory approval. Reliance on third-party providers may
expose us to different risks than if we were to manufacture product
candidates ourselves. The facilities used by our contract
manufacturers to manufacture our product candidates must be
approved by the FDA or other regulatory authorities, pursuant to
inspections that will be conducted after we submit our NDA or
comparable marketing application to the FDA or other regulatory
authority. We do not have control over a supplier’s or
manufacturer’s compliance with these laws, regulations and
applicable cGMP standards and other laws and regulations, such as
those related to environmental health and safety matters. If our
contract manufacturers cannot successfully manufacture material
that conforms to our specifications and the strict regulatory
requirements of the FDA or others, they will not be able to secure
and/or maintain regulatory approval for their manufacturing
facilities. In addition, we have no control over the ability of our
contract manufacturers to maintain adequate quality control (QC),
quality assurance (QA) and qualified personnel. If we are compelled
or we wish to find alternative manufacturing facilities, this could
significantly impact our ability to develop, obtain regulatory
approval for or market our product candidates. Any failure to
achieve and maintain compliance with these laws, regulations and
standards could subject us to the risk that we may have to suspend
the manufacturing of our product candidates or that obtained
approvals could be revoked, which would adversely affect our
business and reputation.
Third-party providers may breach
agreements they have with us because of factors beyond our control.
Contract manufacturers often encounter difficulties involving
production yields, QC and QA, as well as shortages of qualified
personnel. They may also terminate or refuse to renew their
agreements because of their own financial difficulties or business
priorities, potentially at a time that is costly or otherwise
inconvenient for us. If we are unable to find adequate replacement
or another acceptable solution in time, our clinical studies could
be delayed or our commercial activities could be harmed.
In addition, the fact that we are
dependent on our suppliers and other third parties for the
manufacture, storage and distribution of our product candidates
means that we are subject to the risk that our product candidates
and, if approved, commercial products may have manufacturing
defects that we have limited ability to prevent or control. The
sale of products containing such defects could result in recalls or
regulatory enforcement action that could adversely affect our
business, financial condition and results of operations.
Growth in the costs and expenses of
components or raw materials may also adversely influence our
business, financial condition and results of operations. Supply
sources could be interrupted from time to time and, if interrupted,
we cannot be certain that supplies could be resumed (whether in
part or in whole) within a reasonable timeframe and at an
acceptable cost or at all. Our current and anticipated future
dependence upon others for the manufacturing of our current and
future product candidates may adversely affect our future profit
margins and our, or our collaboration partners’, ability to
commercialize any products that receive marketing approval on a
timely and competitive basis.
Our
collaboration arrangements with our strategic partners may make us
an attractive target for potential acquisition under certain
circumstances.
Under certain circumstances, due to
the structure of our collaboration arrangements with our strategic
partners, our strategic partners may prefer to acquire us rather
than pay the milestone payments or royalties under the
collaboration arrangements, which may bring additional
uncertainties to our business development and prospects. For
example, under our collaboration arrangements with Genentech, Lilly
and Janssen, we may become entitled to substantial milestone
payments and royalties. As a result, rather than paying the
milestone payments or royalties, Genentech, Lilly or Janssen, or
one of their affiliates including Roche or Johnson & Johnson,
may choose to acquire us.
Risks related to intellectual
property
We may
not have sufficient patent terms to protect our products and
business effectively.
Patents have a limited lifespan. In
the US, the natural expiration of a patent is generally 20 years
after it is filed. Although various extensions or adjustments may
be available, such as adjustments based on certain delays caused by
the US Patent and Trademark Office (USPTO) the life of a patent,
and the protection it affords, is limited. Given the amount of time
required for the development, testing and regulatory review of new
product candidates, patents protecting such candidates might expire
before or shortly after such candidates are commercialized. As a
result, our owned, co-owned and licensed patent portfolios may not
provide us with sufficient rights to exclude others from
commercializing products similar or identical to ours or otherwise
provide us with a competitive advantage. Even if patents covering
our product candidates are obtained and unchallenged, once the
patent life has expired for a product, we may be open to
competition from generic medications.
Although patent term extensions
under the Hatch-Waxman Act, in the US and under supplementary
protection certificates (SPCs) in Europe may be available to extend
the patent exclusivity term for our products, we cannot provide any
assurances that any such patent term extension will be obtained
and, if so, for how long. The Hatch-Waxman Act permits a patent
extension term of up to 5 years as compensation for patent term
lost during the FDA regulatory review process. A patent term
extension cannot extend the remaining term of a patent beyond a
total of 14 years from the date of product approval, only one
patent may be extended, and only those claims covering the approved
drug, a method for using it or a method for manufacturing it may be
extended. However, we may not be granted any extension because of,
for example, failing to exercise due diligence during the testing
phase or regulatory review process, failing to apply within
applicable deadlines, failing to apply prior to expiration of
relevant patents or otherwise failing to satisfy applicable
requirements. It is not possible to base an SPC in Europe on a
patent in a European Member State if that patent expires before the
Market Authorization (MA) of the clinical product, protected by the
patent, is obtained. As the “product” (active ingredient(s)) must
be “protected by a basic patent in force,” only a granted patent
that is in force, and remains in force until it reaches the end of
its full term, can serve as a “basic patent” upon which an SPC can
be based. Therefore, expired patents and pending patent
applications cannot serve as the basis for an SPC. Given the
relatively long clinical development timelines of biologicals and
new chemical entities for therapeutic purposes, we may not be
granted any patent extensions as we might fail to apply for the
extensions prior to expiration of relevant patents. Moreover, the
applicable time period or the scope of patent protection afforded
could be less than we request. If we are unable to obtain patent
term extension or if the term of any such extension is less than we
request, such result could have a material adverse effect on our
business.
We or
our licensing or collaboration partners may become subject to
intellectual property-related litigation or other proceedings to
protect or enforce our patents or the patents of our licensors or
licensees and collaborators, any of which could be expensive,
time-consuming, and unsuccessful, and may ultimately result in our
loss of ownership of intellectual property.
Competitors may infringe our
patents or the patents of our licensors or collaborators. To
counter such infringement, we may be required to file infringement
claims against those competitors, which can be expensive and
time-consuming. If we or one of our licensing or collaboration
partners were to initiate legal proceedings against a third party
to enforce a patent covering one of our product candidates, the
defendant could counterclaim that the patent covering our product
candidate is invalid or unenforceable or that the defendant’s
products do not infringe our or our licensing collaborators’
patents or that we or our licensing collaborators infringe the
defendant’s patents. In patent litigation in the US, defendant
counterclaims alleging invalidity, unenforceability and
non-infringement are commonplace. Grounds for a validity challenge
could be an alleged failure to meet any of several statutory
requirements, including lack of novelty, obviousness,
obviousness-type double patenting, lack of written description, or
non-enablement. Grounds for an unenforceability assertion could be
an allegation that someone connected with prosecution of the patent
withheld relevant information from the USPTO, or made a misleading
statement, during prosecution. In addition, third parties may raise
similar claims before administrative bodies in the US or abroad,
even outside the context of litigation. Such mechanisms include
re-examination, post-grant review, inter partes review, interference
and derivation proceedings as well as equivalent proceedings in
foreign jurisdictions, such as opposition proceedings in Europe.
The outcome following legal assertions of invalidity and
unenforceability is unpredictable. Such proceedings or patent
litigations could result in the revocation or cancellation of or
amendment to our patents in such a way that they no longer cover
our product candidates or otherwise provide any competitive
advantage. With respect to the validity question, for example, we
cannot be certain that there is no invalidating prior art, of which
the patent examiner and we or our licensing or collaboration
partners were unaware during prosecution. A court may also refuse
to stop a third party from using the technology in question on the
grounds that our patents do not cover that technology. An adverse
result in any proceeding could put one or more of our patents at
risk of being invalidated or interpreted narrowly, which could have
a material adverse effect on our business and financial
condition.
Interference proceedings provoked
by third parties, brought by us or declared by the USPTO may be
necessary to determine the priority of inventions with respect to
our patents or patent applications or those of our licensors,
licensees or collaborators. An unfavorable outcome could require us
or our licensing or collaboration partners to cease using the
related technology or to attempt to license rights to it from the
prevailing party. Our business could be materially harmed if the
prevailing party does not offer us or our licensing or
collaboration partners a license on commercially reasonable terms
or at all. If we or our licensing or collaboration partners are
unsuccessful in any interference proceedings, we may lose our
ownership of intellectual property or our patents may be narrowed
or invalidated. There can be no assurance as to the outcome of the
interference and opposition proceedings, and any of the foregoing
could result in a material adverse effect on our business,
financial condition, results of operations or prospects.
Our defense of litigation,
interference proceedings or other intellectual property-related
proceedings may fail and, even if successful, may result in
substantial costs and distract our management and other employees
from their normal responsibilities. Such litigation or proceedings
could substantially increase our operating losses and could
substantially reduce the funds necessary to continue our clinical
studies and research programs or force us to license necessary
technology from third parties, or enter into development
partnerships that would help us bring our product candidates to
market. We may not be able to prevent, alone or with our licensing
or collaboration partners, misappropriation of our intellectual
property rights, particularly in countries where the laws may not
protect those rights as fully as in the US.
Furthermore, because of the
substantial amount of discovery required in connection with
intellectual property litigation, there is a risk that some of our
confidential information could be compromised by disclosure during
this type of litigation. There could also be public announcements
of the results of hearings, motions, decisions or other interim
proceedings or developments. If securities analysts or investors
perceive these results to be negative, it could have a material
adverse effect on the price of our common shares.
If we or
our licensing or collaboration partners are unable to obtain and
maintain effective patent rights for our technologies, product
candidates or any future product candidates, or if the scope of the
patent rights obtained is not sufficiently broad, our competitors
could develop and commercialize products and technology similar or
identical to ours, and our, or our collaboration partners’ ability
to successfully commercialize our products and technology may be
adversely affected.
We rely upon a combination of
patents, trade secret protection and confidentiality agreements to
protect the intellectual property related to our technologies and
product candidates. Our success depends in large part on our and
our licensing or collaboration partners’ ability to obtain and
maintain patent and other intellectual property protection in the
US, the EU and other countries with respect to our proprietary
technologies and product candidates. In particular, Genentech,
Lilly, Janssen or our other licensing or collaboration partners may
be dependent on a license with a third party for the development
and future commercialization of our product candidates. If such
license is not granted or is terminated, Genentech, Lilly, Janssen
or other licensing or collaboration partners may be required to
cease development and commercialization of our product candidates,
any of which could have a material adverse effect on our business,
financial condition, results of operations or prospects.
We have sought to protect our
proprietary position by filing patent applications in the US and
abroad related to any of our novel technologies and products that
are important to our business. This process is expensive,
time-consuming, and complex, and we may not be able to file and
prosecute all necessary or desirable patent applications at a
reasonable cost, in a timely manner or in all jurisdictions. It is
also possible that we will fail to identify patentable aspects of
our or our licensing or collaboration partners’ research and
development output before it is too late to obtain patent
protection. Moreover, in some circumstances, we do not have the
right to control the preparation, filing and prosecution of patent
applications, or to maintain the patents, covering technology that
we license to or from third parties. Therefore, these patents and
applications may not be prosecuted and enforced in a manner
consistent with the best interests of our business.
The patent position of
pharmaceutical and biopharmaceutical companies generally is highly
uncertain and involves complex legal and factual questions for
which legal principles remain unsolved. As a result, the
inventorship, issuance, scope, validity, enforceability and
commercial value of our patent rights are highly uncertain. The
pending or future patent applications that we own, co-own or
in-license may fail to issue, fail to result in issued patents with
claims that cover our product candidates in the US or in other
foreign countries, or fail to effectively prevent others from
commercializing competitive technologies and product candidates.
Changes in either the patent laws or interpretation of the patent
laws in the US and other countries may diminish the value of our
patents or narrow the scope of our patent protection.
We may not be aware of all
third-party intellectual property rights potentially relating to
our technologies or product candidates. Publications of discoveries
in the scientific literature often lag behind the actual
discoveries, and patent applications in the US and other
jurisdictions remain confidential for a period of time after
filing, and some remain so until issued. Therefore, we cannot be
certain that we were the first to file any patent application
related to our product candidates or technologies, or whether we
were the first to make the inventions claimed in our owned or
co-owned patents or pending patent applications, nor can we know
whether those from whom we license patents were the first to make
the inventions claimed or were the first to file.
There is no assurance that all
potentially relevant prior art relating to our patents and patent
applications has been found, which can invalidate a patent or
prevent a patent from issuing from a pending patent application.
Even if patents do successfully issue, and even if such patents
cover our product candidates, third parties may challenge their
validity, enforceability, or scope, which may result in such
patents being narrowed, found unenforceable or invalidated, which
could allow third parties to commercialize our technology or
products and compete directly with us, without payment to us, or
result in our or our collaboration partners’ inability to
manufacture or commercialize products without infringing
third-party patent rights. Furthermore, even if they are
unchallenged, our patents and patent applications may not
adequately protect our intellectual property, provide exclusivity
for our product candidates, prevent others from designing around
our claims or provide us with a competitive advantage. Any of these
outcomes could impair our ability to prevent competition from third
parties, which may have a material adverse effect on our
business.
We may
be subject to claims challenging the inventorship of our patents
and other intellectual property.
We may be subject to claims that
former employees, collaborators or other third parties have an
interest or title in our patents or other intellectual property as
an inventor or co-inventor. For example, we may have inventorship
disputes arise from conflicting obligations of consultants, CROs,
CMOs, academic institutions or others who are involved in
developing our product candidates. Litigation may be necessary to
defend against these and other claims challenging inventorship or
our ownership of our patents or other intellectual property. If we
fail in defending any such claims, in addition to paying monetary
damages, we may lose valuable intellectual property rights, such as
exclusive ownership of, or the right to use, valuable intellectual
property. Such an outcome could have a material adverse effect on
our business. Even if we are successful in defending against such
claims, litigation could result in substantial costs and be a
distraction to management and other employees.
Patent
policy and rule changes could increase the uncertainties and costs
surrounding the prosecution of our patent applications and the
enforcement or defense of our issued patents, thereby impairing our
ability to protect our technologies and products.
Changes in either the patent laws
or interpretation of the patent laws in the US, EU or elsewhere
could increase the uncertainties and costs surrounding the
prosecution of patent applications and the enforcement or defense
of issued patents. Assuming the other requirements for
patentability are met, in the US prior to March 15, 2013, the first
to make the claimed invention is entitled to the patent, whereas
outside the US, the first to file a patent application was entitled
to the patent. After March 15, 2013, under the Leahy-Smith America
Invents Act (the Leahy-Smith Act), enacted on September 16, 2011,
the US has moved to a first-to-file system. Under a first-to-file
system, assuming the other requirements for patentability are met,
the first inventor to file a patent application generally will be
entitled to the patent on an invention regardless of whether a
third party was the first to invent the invention. The Leahy-Smith
Act also includes a number of significant changes that affect the
way patent applications are prosecuted and may also affect patent
litigation. These include allowing third-party submission of prior
art to the USPTO during patent prosecution and additional
procedures to attack the validity of a patent by the USPTO
administered during post grant proceedings, including
re-examination proceedings, inter
partes review, post-grant review and derivation proceedings.
Therefore, the Leahy-Smith Act and its implementation increases the
uncertainties and costs surrounding the prosecution of our patent
applications and the enforcement or defense of our issued patents,
all of which could have a material adverse effect on our business,
financial condition, results of operations and prospects. In
addition, future actions by the US Congress, the federal courts and
the USPTO could cause the laws and regulations governing patents to
change in unpredictable ways. Any of the foregoing could harm our
business, financial condition and results of operations.
In addition, the patent positions
of companies in the development and commercialization of biologics
and pharmaceuticals are particularly uncertain. US Supreme Court
rulings have narrowed the scope of patent protection available in
certain circumstances and weakened the rights of patent owners in
certain situations. This combination of events has created
uncertainty with respect to the validity and enforceability of
patents, once obtained. Depending on future actions by the US
Congress, the federal courts, and the USPTO, the laws and
regulations governing patents could change in unpredictable ways
that could have a material adverse effect on our existing patent
portfolio and our ability to protect and enforce our intellectual
property in the future in the US.
If we
are unable to maintain effective proprietary rights for our
technologies, product candidates or any future product candidates,
we may not be able to compete effectively in our markets.
In addition to the protection
afforded by patents, we rely on trade secret protection and
confidentiality agreements to protect proprietary know-how that is
not patentable or that we elect not to patent, processes for which
patents are difficult to enforce, and any other elements of our
product candidate discovery and development processes that involve
proprietary know-how, information or technology that is not covered
by patents. However, trade secrets can be difficult to protect and
some courts inside and outside the US are less willing or unwilling
to protect trade secrets. The EU has introduced a Directive on
trade secrets increasing the standards for protection. Because we
rely on our advisors, employees and third-party contractors and
consultants to research and develop and to manufacture our product
candidates, we must, at times, share our intellectual property with
them. We seek to protect our intellectual property and other
proprietary technology in part by entering into confidentiality
agreements and master service agreements, if applicable, material
transfer agreements, consulting agreements or other similar
agreements with our advisors, employees, contractors, consultants,
licensing and collaboration partners, and other third parties with
confidentiality provisions. These agreements typically limit the
rights of these third parties to use or disclose our confidential
information, including our intellectual property and trade secrets.
These agreements also typically restrict the ability of third
parties to publish data potentially relating to our intellectual
property, although our agreements may contain certain limited
publication rights. For example, any academic institution that we
may collaborate with in the future may expect to be granted rights
to publish data arising out of such collaboration, provided that we
may have the right to be notified in advance and given the
opportunity to delay publication for a limited time period in order
for us to secure patent protection of intellectual property rights
arising from the collaboration, in addition to the opportunity to
remove confidential or trade secret information from any such
publication. We also conduct joint research and development
programs that may require us to share intellectual property under
the terms of our research and development or similar agreements.
However, we cannot guarantee that we have entered into such
agreements with each party that may have or have had access to our
trade secrets or other confidential information or proprietary
technology and processes, or that such agreements will not be
breached or that our trade secrets or other confidential
information will not otherwise be disclosed. Despite the
contractual provisions employed when working with these advisors,
employees and third-party contractors and consultants, the need to
share intellectual property and other confidential information
increases the risk that such confidential information becomes known
by our competitors, is inadvertently incorporated into the product
development of others or is disclosed or used in violation of these
agreements. Additionally, our grant agreements typically provide
for dissemination of results to academic institutions and to the
general public. As a result, our information may be disseminated
with the loss of protection status.
We also seek to preserve the
integrity and confidentiality of our data and trade secrets by
maintaining the physical security of our premises and the physical
and electronic security of our information technology systems.
Despite our efforts to protect our intellectual property, our
competitors may discover our trade secrets through breach of our
agreements by third parties, for which we may not have adequate
remedies for any breach, or publication of information by any of
our CROs, academic partners, funding organizations or our licensing
or collaboration partners. Additionally, if the steps we take or
that we impose on our CROs maintain our trade secrets are deemed
inadequate by law, we may have insufficient recourse against third
parties for misappropriating such trade secrets. Misappropriation
or unauthorized disclosure of our trade secrets could impair our
competitive position and may have a material adverse effect on our
business. Moreover, if any of our trade secrets were to be lawfully
obtained or independently developed by a competitor or other third
party, we would have no right to prevent such competitor or other
third party from using that technology or information to compete
with us. A competitor’s or other third party’s discovery of our
intellectual property would impair our competitive position and
have a material adverse effect on our business.
Further, the laws of some foreign
countries do not protect proprietary rights to the same extent or
in the same manner as the laws of the US. As a result, we may
encounter significant problems in protecting and defending our
intellectual property both in the US and abroad. If we are unable
to prevent material disclosure of the intellectual property related
to our technologies to third parties, we will not be able to
establish or maintain a competitive advantage in our market, which
could materially adversely affect our business, financial condition
and results of operations.
Despite confidentiality clauses
within our employment agreements, we cannot ensure that departing
employees will not breach any post-termination commitments in such
agreements by allowing others to access our trade secrets.
Obtaining and maintaining our patent protection depends on
compliance with various procedural, document-submission,
fee-payment and other requirements imposed by governmental patent
agencies, and our patent protection could be reduced or eliminated
for noncompliance with these requirements.
Periodic maintenance fees, renewal
fees, annuity fees and various other government fees on a patent
and patent application are due to be paid to the USPTO and foreign
patent agencies in several stages over the lifetime of the patent
and patent application. The USPTO and various foreign governmental
patent agencies require compliance with a number of procedural,
documentary, fee-payment and other similar provisions during the
patent application process. We employ reputable law firms and other
professionals to help us comply with these requirements and we are
also dependent on our licensors or collaboration partners to take
the necessary action to comply with these requirements with respect
to certain of our intellectual property. Although an inadvertent
lapse can in many cases be cured by payment of a late fee or by
other means in accordance with the applicable rules, there are
situations in which noncompliance can result in abandonment or
lapse of the patent or patent application, resulting in partial or
complete loss of patent rights in the relevant jurisdiction.
Noncompliance events that could result in abandonment or lapse of a
patent or patent application include, but are not limited to,
failure to respond to official actions within prescribed time
limits, nonpayment of fees and failure to properly legalize and
submit formal documents. In such an event, our competitors might be
able to enter the market, which would have a material adverse
effect on our business.
The
patent protection and patent prosecution for some of our product
candidates is dependent on third parties.
Although we normally seek to
obtain the right to control prosecution, maintenance and
enforcement of the patents relating to our product candidates,
there may be times when the filing and prosecution activities for
patents relating to our product candidates are controlled by our
licensors or collaboration partners. If any of our current or
future licensing or collaboration partners fail to prosecute,
maintain and enforce such patents and patent applications in a
manner consistent with the best interests of our business,
including by payment of all applicable fees for patents covering
our product candidates, we could lose our rights to the
intellectual property or our exclusivity with respect to those
rights, our or our collaboration partners’ ability to develop and
commercialize those product candidates may be adversely affected
and we may not be able to prevent competitors from making, using,
and selling competing products. In addition, even where we have the
right to control patent prosecution of patents and patent
applications we have licensed to and from third parties, we may
still be adversely affected or prejudiced by actions or inactions
of our licensees, our licensors and their counsel that took place
prior to the date upon which we assumed control over patent
prosecution.
Additionally, we may be adversely
affected or prejudiced by actions or inactions of our external and
internal patent counsels working solely on our projects or our
joint patent counsels representing us and our collaboration
partners.
If we
fail to comply with the obligations in our intellectual property
agreements, including those under which we license intellectual
property and other rights to or from third parties, or otherwise
experience disruptions to our business relationships with our
licensees, our licensors and collaboration partners, we could lose
intellectual property rights that are important to our
business.
We are a party to a number of
intellectual property license and co-ownership agreements and
research and development collaborations that are important to our
business and expect to enter into additional such agreements in the
future. Under certain circumstances, the royalties payable to us
under these agreements are subject to certain reductions, which may
have a materially adverse effect on our business, financial
condition, results of operations and prospects. In addition, our
existing agreements impose, and we expect that future agreements
will impose, various diligence, commercialization, milestone
payment, royalty and other obligations on us. If we fail to comply
with our obligations under these agreements, we may be required to
make certain payments to the licensor, we may lose the exclusivity
of our license or the licensor may have the right to terminate the
license, in which event we would not be able to develop or market
products covered by the license.
Licensing of intellectual property
is of critical importance to our business and involves complex
legal, business and scientific issues. Disputes may arise regarding
intellectual property subject to a licensing or co-ownership
agreement, including:
|
• |
the scope of rights granted under the agreement, any
restrictions in licensed fields and other interpretation-related
issues;
|
|
• |
the extent to which our technology and processes infringe or
otherwise violate the intellectual property of the licensor, the
licensee or partner that is not subject to the agreement;
|
|
• |
the sublicensing of patent and other rights;
|
|
• |
the diligence, development and commercialization obligations
under the agreement and what activities satisfy those
obligations;
|
|
• |
the ownership of inventions and know-how resulting from the
joint or mutual creation or use of intellectual property by our
licensors or collaboration partners and us;
|
|
• |
the priority of invention in patented technology;
|
|
• |
non-compete commitments; and
|
|
• |
consequences for changes in control.
|
If disputes over intellectual
property and other rights that we own, have licensed or co-own
prevent or impair our ability to maintain our current licensing or
exclusivity arrangements on acceptable terms, we or our
collaboration partners may be unable to successfully develop and
commercialize the affected product candidates.
In addition, certain provisions in
the agreements under which we currently license intellectual
property or technology to and from third parties may be susceptible
to multiple interpretations. The resolution of any contract
interpretation disagreement that may arise could narrow what we
believe to be the scope of our rights to the relevant intellectual
property or technology, increase what we believe to be our
financial or other obligations under the relevant agreement, or
decrease the third party’s financial or other obligations under the
relevant agreement, any of which could have a material adverse
effect on our business, financial condition, results of operations
and prospects.
We or
our licensors or licensees and collaborators may not be successful
in obtaining or maintaining necessary rights to our product
candidates through acquisitions and in-licenses.
Our or our licensors or licensees
and collaborators programs may require the use of proprietary
rights held by third parties in the future, and the growth of our
business will likely depend in part on our ability to acquire,
in-license, maintain or use these proprietary rights. In addition,
our product candidates may require specific processes and/or
formulations to work effectively and efficiently and the rights to
these processes and/or formulations may be held by others. We or
our licensors or licensees may be unable to acquire or in-license
from third parties any compositions, methods of use, processes, or
other third-party intellectual property rights that we identify as
necessary for our product candidates. The licensing and acquisition
of third-party intellectual property rights is a competitive area,
and a number of more established companies may pursue strategies to
license or acquire third-party intellectual property rights that we
may consider attractive or necessary. These established companies
may have a competitive advantage over us due to their size, cash
resources, and greater clinical development and commercialization
capabilities. In addition, companies that perceive us to be a
competitor may be unwilling to assign or license rights to us. We
or our licensors or licensees also may be unable to license or
acquire third-party intellectual property rights on terms that
would allow us to make an appropriate return on our
investment.
For example, we sometimes
collaborate with US and foreign academic institutions to accelerate
our preclinical research or development under written agreements
with these institutions. Typically, these institutions provide us
with an option to negotiate a license to any of the institution’s
rights in technology resulting from the collaboration. Regardless
of such option, we may be unable to negotiate a license within the
specified timeframe or under terms that are acceptable to us. If we
are unable to do so, the institution may offer the intellectual
property rights to other parties, potentially blocking our ability
to pursue our applicable product candidate or program.
If we are unable to successfully
obtain a license to third-party intellectual property rights
necessary for the development of a product candidate or program, we
may have to abandon development of that product candidate or
program and our business and financial condition could
suffer.
Third-party claims of intellectual property infringement may expose
us to substantial liability or may prevent or delay our or our
collaboration partners’ development and commercialization
efforts.
Numerous US- and foreign-issued
patents and pending patent applications, which are owned by third
parties, exist in the fields in which we are developing product
candidates. For example, we are aware of third-party patents or
patent applications that may be construed to cover one or more of
our product candidates. If these patents are asserted against us or
our licensing or collaboration partners and either we or our
licensing or collaboration partners are found to infringe any of
these patents, and are unsuccessful in demonstrating that such
patents are invalid or unenforceable, then we and our licensing or
collaboration partners could be required to pay substantial
monetary damages or cease further development or commercialization
of one or more of our product candidates or be compelled to enter
into onerous licenses with such third parties. There may also be
other third-party patents or patent applications with claims to
materials, formulations, methods of manufacture or methods of
treatment related to the use or manufacture of our product
candidates and technology. Although we generally conduct a
freedom-to-operate search and review with respect to our product
candidates, we cannot guarantee that our search and review is
complete and thorough, nor can we be sure that we have identified
each and every patent and pending application in the US and abroad
that is relevant or necessary to the manufacturing or
commercialization of our product candidates or use of our
technology. Because patent applications can take many years to
issue, there may be currently pending patent applications that may
later result in issued patents that our product candidates may
infringe. In addition, third parties may file and obtain additional
patents in the future and claim that use of our technologies
infringes upon these patents.
Third parties may assert
infringement claims against us based on existing patents or on
patents that may be granted in the future, regardless of merit.
Even if we believe such claims are without merit, a court of
competent jurisdiction could hold that these third-party patents
are valid, enforceable and infringed, which could materially and
adversely affect our or our collaboration partners’ ability to
commercialize our product candidates or technologies covered by the
asserted third-party patents.
Parties making claims against us
may also obtain injunctive or other equitable relief, which could
effectively block our or our collaboration partners’ ability to
further develop and commercialize one or more of our product
candidates. Defense of these claims, regardless of their merit,
would involve substantial litigation expense and would be a
substantial diversion of management and employee resources from our
business. In the event of a successful claim of infringement
against us, we may have to pay substantial damages, including
treble damages and attorneys’ fees for willful infringement, pay
royalties, redesign our infringing products or obtain one or more
licenses from third parties, which may be impossible or require
substantial time and monetary expenditure. Any of the foregoing
could have a material and adverse effect on our business, financial
conditions, results of operations and prospects.
In addition, claims that we have
misappropriated the confidential information or trade secrets of
third parties could have a similar negative impact on our business,
financial condition, results of operations and prospects.
There could also be public
announcements of the results of hearings, motions, decisions, or
other interim proceedings or developments. If securities analysts
or investors perceive these results to be negative, it could have a
material adverse effect on the price of our common shares.
Some of our competitors may have
substantially greater resources and more mature and developed
intellectual property portfolios than we do, and may be able to
sustain the costs of complex intellectual property litigation to a
greater degree and for longer periods of time than we could. In
addition, patent-holding companies that focus solely on extracting
royalties and settlements by enforcing patent rights may target us.
As the pharmaceutical and biopharmaceutical industries expand and
more patents are issued, the risk increases that our product
candidates may be subject to claims of infringement of the patent
rights of third parties. The uncertainties resulting from the
initiation and continuation of patent litigation or other
proceedings could have a material adverse effect on our ability to
compete in the marketplace.
We may be
subject to claims that our employees, consultants, or independent
contractors have wrongfully used or disclosed confidential
information of third parties or that our employees have wrongfully
used or disclosed alleged trade secrets of their former
employers.
We employ and utilize the services
of individuals who were previously employed or provided services to
universities or other pharmaceutical or biopharmaceutical
companies, including our competitors or potential competitors.
Although we try to ensure that our employees, consultants, and
independent contractors do not use the proprietary information or
know-how of others in their work for us, we may be subject to
claims that we or our employees, consultants or independent
contractors have inadvertently or otherwise used or disclosed
intellectual property, including trade secrets or other proprietary
information, of any of our employees’, consultants’ or independent
contractors’ former employers or of other third parties. Litigation
may be necessary to defend against these claims. If we fail in
defending any such claims, in addition to paying monetary damages,
we may lose valuable intellectual property rights or personnel,
which could have a material adverse effect on our business. Even if
we are successful in defending against such claims, litigation
could result in substantial costs and be a distraction to
management and other employees.
In addition, although it is our
policy to require our employees, consultants and independent
contractors who may be involved in the conception or development of
intellectual property to execute agreements assigning such
intellectual property to us, we may be unsuccessful in executing
such an agreement with each party who, in fact, conceives or
develops intellectual property that we regard as our own. The
assignment of intellectual property rights may not be
self-executing or the assignment agreements may be breached, and we
may be forced to bring claims against third parties, or defend
claims that they may bring against us, to determine the ownership
of what we regard as our intellectual property.
We may
not be able to protect our intellectual property rights throughout
the world.
Filing, prosecuting and defending
patents on product candidates in all countries throughout the world
would be prohibitively expensive, and our intellectual property
rights in some countries outside the US may be less extensive than
those in the US. In addition, the laws of some foreign countries do
not protect intellectual property rights to the same extent as the
laws in the US. Consequently, we may not be able to prevent third
parties from practicing our inventions in all countries outside the
US, or from selling or importing products made using our inventions
in and into the US or other jurisdictions. In the ordinary course
of prosecution and maintenance activities, we determine whether to
seek patent protection outside the US and in which countries. This
also applies to patents we have acquired or in-licensed from third
parties. In some cases, we, or our predecessors in interest or
licensors of patents within our portfolio, have sought patent
protection in a limited number of countries for patents covering
our product candidates. Competitors may use our technologies and
products in jurisdictions where we have not obtained or are unable
to adequately enforce patent protection to develop their own
products and further, may export otherwise infringing products to
territories where we have patent protection but enforcement is not
as strong as that in the US. These products may compete with our
products and our patents or other intellectual property rights may
not be effective or sufficient to prevent them from competing,
which would have a material adverse effect on our business and
financial positions.
Many companies have encountered
significant problems in protecting and defending intellectual
property rights in foreign jurisdictions. The legal systems of
certain countries, particularly certain developing countries, do
not favor the enforcement of patents, trade secrets, and other
intellectual property protection, particularly those relating to
biotechnology products, which could make it difficult for us to
stop the infringement, misappropriation or other violations of our
intellectual property and proprietary rights. Proceedings to
enforce our patent rights in foreign jurisdictions, whether or not
successful, could result in substantial costs and divert our
efforts and attention from other aspects of our business, could put
our patents at risk of being invalidated or interpreted narrowly
and our patent applications at risk of not issuing, and could
provoke third parties to assert claims against us. We may not
prevail in any lawsuits that we initiate and the damages or other
remedies awarded, if any, may not be commercially meaningful.
Accordingly, our efforts to enforce our intellectual property
rights around the world may be inadequate to obtain a significant
commercial advantage from the intellectual property that we develop
or license.
If our
trademarks and trade names are not adequately protected, then we
may not be able to build name recognition in our markets of
interest, our names and brands may be misappropriated by third
parties, and our business may be adversely affected
We have filed trademark
applications seeking protection for our corporate name, logo,
Nasdaq Global Market symbol and selected names of our technology
platforms in selected geographies. While we have been granted
registrations in certain geographies for certain trademarks, there
is no guarantee that our trademark applications will be approved by
the respective authorities at all or that we will not be required
to narrow the scope of protection in certain or all geographies.
Our applications may face opposition from third parties,
potentially resulting in the lack of protection or narrower
protection. Our trademarks or trade names may be challenged,
infringed, circumvented, declared generic or determined to be
infringing on other marks. We may not be able to protect our rights
to these trademarks and trade names, which we need to build name
recognition among potential partners or customers in our markets of
interest. At times, competitors or other third parties may adopt
trade names, domain names or trademarks similar to ours, thereby
impeding our ability to build brand identity and possibly leading
to market confusion. In addition, there could be potential trade
name or trademark infringement claims brought by owners of other
registered trademarks or trademarks that incorporate variations of
our trademarks or trade names. Over the long term, if we are unable
to establish name recognition based on our trademarks and trade
names, then we may not be able to compete effectively and our
business may be adversely affected. Our efforts to enforce or
protect our proprietary rights related to trademarks and domain
names may be ineffective and could result in substantial costs and
diversion of resources, and could adversely affect our business,
financial condition, results of operations and growth
prospects.
Risks related to our financial
condition and capital requirements
We are a
clinical-stage company and have a history of operating losses. We
anticipate that we will continue to incur losses
for the foreseeable future.
We are a clinical-stage
biopharmaceutical company. Since 2003, although we have received
upfront and milestone payments from our collaboration partners and
certain other contract revenue, we have also incurred significant
operating losses. We incurred net losses (defined as net loss
attributable to owners of the Company) of CHF 73.0 million for the
year ended December 31, 2021. In addition, we had accumulated
losses of CHF 200.9 million as of December 31, 2021.
Our losses have resulted
principally from research and development expenses and from general
business and administrative expenses. We expect to continue to
incur significant operating losses in the future as we continue our
research and development efforts for our current and future product
candidates and seek to obtain regulatory approval and
commercialization of such product candidates.
To date, the Company has financed
its liquidity requirements primarily from its public offerings,
share issuances, contract revenues from license and collaboration
agreements and grants. We have no products approved for
commercialization and have never generated any revenues from
product sales. Biopharmaceutical and pharmaceutical product
development is a highly speculative undertaking and involves a
substantial degree of risk. It may be several years, if ever,
before we or our collaboration partners complete pivotal clinical
studies and have a product candidate approved for commercialization
and we begin to generate revenue or royalties from product
sales.
Although
we have generated revenues from upfront and milestone payments
related to our license and collaboration agreements, we have never
generated any revenue from product sales and may never be
profitable.
Although we have generated contract
revenue from upfront and milestone payments related to our license
and collaboration agreements, we have no products approved for
commercialization and have never generated any revenue from product
sales. Our ability to generate revenue and achieve profitability
depends on our and our licensors’ and collaboration partners’
ability to successfully complete the development of, and obtain the
marketing approvals necessary, to commercialize one or more of our
product candidates. We do not anticipate generating revenue from
product sales unless and until we or our collaboration partners
obtain regulatory approval for, and commercialize, our product
candidates. Our ability to generate future revenue from product
sales depends heavily on our and our collaboration partners’
success in many areas, including but not limited to:
|
• |
successfully completing research and clinical development of
our product candidates, by us or our collaboration partners, as the
case may be;
|
|
• |
obtaining marketing approvals for our clinical product
candidates, including ACI-35, ACI-24 for AD and DS, ACI-7104,
semorinemab, crenezumab, Morphomer Tau, PI-2620 and our a-syn PET
tracer, for which we or our collaboration partners complete
clinical studies;
|
|
• |
developing a sustainable and scalable manufacturing process
for any approved product candidates, and maintaining supply and
manufacturing relationships with third parties that can conduct the
process and provide adequate (in amount, quality and time) products
to support clinical development and the market demand for our
product candidates, if approved;
|
|
• |
launching and commercializing product candidates for which we
obtain marketing approval, either directly or with a collaborator
or distributor;
|
|
• |
obtaining market acceptance of our product candidates as
viable treatment or diagnostic options;
|
|
• |
addressing any competing technological and market
developments;
|
|
• |
identifying, assessing, acquiring and/or developing new
product candidates;
|
|
• |
negotiating favorable terms in any collaboration, licensing,
or other similar arrangements into which we may enter;
|
|
• |
maintaining, protecting, acquiring and expanding our portfolio
of intellectual property rights, including patents, trade secrets
and know-how; and
|
|
• |
attracting, hiring and retaining qualified personnel.
|
Because of the numerous risks and
uncertainties with biopharmaceutical and pharmaceutical product
development, we are unable to accurately predict the timing or
amount of increased expenses and when, or if, we will be able to
achieve profitability. Our expenses could increase beyond
expectations if we are required by the FDA, the EMA or other
regulatory agencies, domestic or foreign, to change our
manufacturing processes, or to perform clinical, nonclinical or
other types of studies in addition to those that we currently
anticipate. In cases where we are successful in obtaining
regulatory approvals to market one or more of our product
candidates, our revenue will be dependent, in part, upon the size
of the markets in the territories for which we gain regulatory
approval, the accepted price for the product, the ability to obtain
coverage and reimbursement at any price, and whether we own the
commercial rights for that territory. If the number of our
addressable patients is not as significant as we estimate, the
indication approved by regulatory authorities is narrower than we
expect, the treatment population is narrowed by competition,
physician choice or treatment guidelines or other commercial
related factors we may not generate significant revenue from sales
of such products, even if approved. Accordingly, we may not be
profitable in the future from the sale of any approved
products.
We or our collaboration partners
may be unable to develop and commercialize any of our current or
future product candidates and, even if we do, may not achieve
profitability in the future. Even if we do achieve profitability in
the future, we may not be able to sustain or increase profitability
on a quarterly or annual basis. Our failure to be profitable in the
future would decrease the value of our company and could impair our
ability to raise capital, expand our business or continue our
operations. A decline in the value of our company could cause you
to lose all or part of your investment.
If we
fail to obtain additional funding, we may delay, reduce or
eliminate our product development programs or commercialization
efforts.
We are currently advancing our
clinical product candidates through clinical development, either
together with a collaboration partner (ACI-35, semorinemab,
Morphomer Tau, crenezumab and PI-2620) or independently (ACI-7104,
ACI-24 for AD and for DS and our a-syn PET tracer). We expect our
research and development expenses to continue to increase in
connection with our ongoing activities, particularly as we and/or
our collaboration partners continue our ongoing studies and
initiate new studies of ACI-35, ACI-24 for AD and DS, ACI-7104,
Morphomer Tau, PI-2620 and our a-syn PET tracer and initiate
preclinical and clinical development of our other product
candidates.
As of December 31, 2021, we had
cash and cash equivalents of CHF 82.2 million and short-term
financial assets of CHF 116.0 million resulting in a total
liquidity position of CHF 198.2 million. We currently believe that
our existing capital resources, not including potential milestone
payments, will be sufficient to meet our projected operating
requirements through at least Q1 2024. We have based this estimate
on assumptions that may prove to be wrong, and we could exhaust our
capital resources sooner than we currently expect. In addition,
changing circumstances may cause us to adjust our projected
spending to amounts more than currently expected. We may also need
to raise additional funds sooner than we anticipate due to various
factors such as the scope and rate of progress of our development
activities, regulatory approval outcomes and emergence of competing
technologies, among others.
We expect that we will require
additional capital to develop and commercialize certain of our
product candidates. If we receive regulatory approval for our
current and future product candidates, and if we have not already
licensed such product candidate to a collaboration partner and
choose to commercialize such product candidate independently, we
expect to incur significant commercialization expenses related to
product manufacturing, sales, marketing, distribution and
establishing a regulatory structure, depending on where we choose
to commercialize. Additional funds may not be available on a timely
basis, on favorable terms, or at all, and such funds, if raised,
may not be sufficient to enable us to continue to implement our
long-term business strategy. Additionally, we may be dependent on
the status of the capital markets at the time such capital is
sought. If we are not able to raise capital when needed, we could
be forced to delay, reduce or eliminate our product development
programs or commercialization efforts.
Raising
additional capital may cause dilution to our shareholders, restrict
our operations or require us to relinquish rights to our
intellectual property or future revenue streams.
Until such time, if ever, as we can
generate substantial product royalty revenue, we expect to finance
our liquidity needs through a combination of equity offerings, debt
financings, grants, and license and development agreements in
connection with collaborations. In September 2020, the Company
established an “at the market offering” (ATM) for the sale of up to
USD 80 (CHF 73.9) million worth of our common shares from time to
time by entering into an Open Market Sale Agreement (Sales
Agreement) with Jefferies LLC (Jefferies). In Q2 2021, we filed a new registration
statement on Form F-3 and entered into a new Sales Agreement to
replace and extend the ATM program. We do not have any
material committed external source of funds. In the event we need
to seek additional funds, we may raise additional capital through
the sale of equity, convertible debt or other securities. In such
an event, your ownership interest will be diluted, and the terms of
these securities may include liquidation or other preferences that
adversely affect your rights as a holder of our common shares. Debt
financing, if available, may involve agreements that include
covenants limiting or restricting our ability to take specific
actions, such as incurring additional debt, making capital
expenditures or proposing dividends to our shareholders.
If we raise additional funds
through collaborations, strategic alliances, or marketing,
distribution or licensing arrangements with third parties, we may
have to grant or otherwise relinquish valuable rights to our
intellectual property or future revenue streams.
Our
ability to use tax loss carry-forwards in Switzerland may be
limited.
As of December 31, 2021, we
reported tax loss carry-forwards from financial years 2015 until
2021 for purposes of Swiss corporate income tax in the aggregate
amount of CHF 197.2 million, which could be available to offset
future taxable income. If not used, these tax losses will expire 7
years after the year in which they were incurred. Due to our
limited income, there is a high risk that the tax loss
carry-forwards will expire partly or entirely and we will not be
able to use them to offset future taxable income thereafter for
Swiss corporate income tax purposes.
Exchange
rate fluctuations may materially affect our results of operations
and financial condition.
Under our existing agreements, we
receive and make a significant amount of payments in Swiss Franc,
USD and EUR. As a result, changes and fluctuations in currency
exchange rates between the Swiss Franc and other currencies,
especially the USD and EUR, could have a materially adverse effect
on our operating results. As our reporting currency is the Swiss
Franc, financial line items are converted into Swiss Francs at the
applicable exchange rates. We also expect that in the future, a
significant portion of our revenues and expenses will be
denominated in Swiss Franc, USD and EUR. Therefore, unfavorable
developments in the value of the Swiss Franc as compared to the USD
and EUR or any other currency could have a material adverse effect
on our business, financial condition and results of
operations.
Our
significant in-process research and development (IPR&D) asset
may become impaired.
Our consolidated balance sheet
contains a material IPR&D asset. For an IPR&D asset, the
risk of failure is significant, and there can be no certainty that
the asset will become a successful candidate. Our ability to
realize value on this significant investment is often contingent
upon, among other things, regulatory approvals and market
acceptance. As such, this IPR&D may become impaired and/or be
written off at some time in the future if the associated R&D
effort is abandoned or is curtailed.
Risks related to the regulatory
environment
We
cannot give any assurance that any of our product candidates will
receive regulatory approval, which is necessary before they can be
commercialized.
Our future success is dependent on
our and our collaboration partners’ ability to successfully
develop, obtain regulatory approval for, and then successfully
commercialize one or more product candidates. We currently have two
product candidates that have completed Phase 2 clinical studies and
three that are in a Phase 2 clinical study. We are not permitted to
market or promote any of our product candidates before we receive
regulatory approval from the FDA, EMA or comparable foreign
regulatory authorities, and we may never receive such regulatory
approval for any of our product candidates.
We cannot be certain that any of
our product candidates will be successful in clinical studies or
receive regulatory approval. Applications for our product
candidates could fail to receive regulatory approval for many
reasons, including but not limited to the following:
|
• |
the FDA, EMA or comparable foreign regulatory authorities may
disagree with the design or implementation of our clinical
studies;
|
|
• |
the population studied in the clinical program may not be
sufficiently broad or representative to assure safety in the full
population for which we seek approval;
|
|
• |
the FDA, EMA or comparable foreign regulatory authorities may
disagree with our interpretation of data from nonclinical or
clinical studies;
|
|
• |
the data collected from clinical studies of our product
candidates may not be sufficient to support the submission of an
NDA or other submission or to obtain regulatory approval in the US
or elsewhere;
|
|
• |
we may be unable to demonstrate to the FDA, EMA or comparable
foreign regulatory authorities that a product candidate’s
benefit-risk ratio for its proposed indication is acceptable;
|
|
• |
the FDA, EMA or other regulatory authorities may fail to
approve the manufacturing processes, test procedures and
specifications, or facilities of third-party manufacturers with
which we contract for clinical and commercial supplies; and
|
|
• |
the approval policies or regulations of the FDA, EMA or
comparable foreign regulatory authorities may change significantly
in a manner rendering our clinical data insufficient for
approval.
|
We generally plan to seek
regulatory approval to commercialize our product candidates in the
US, the EU and in additional foreign countries where we have
commercial and typically intellectual property rights. To obtain
regulatory approval in other countries, we must comply with
numerous and varying regulatory requirements of such other
countries regarding safety, efficacy, chemistry, manufacturing and
controls, clinical studies, commercial sales, pricing, marketing
and distribution of our product candidates. Even if we are
successful in obtaining approval in one jurisdiction, we cannot
ensure that we will obtain approval in any other jurisdictions.
Failure to obtain marketing authorization for our product
candidates will result in our being unable to market and sell such
products, which would materially adversely affect our business,
financial condition and results of operations. If we fail to obtain
approval in any jurisdiction, the geographic market for our product
candidates could be limited. Similarly, regulatory agencies may not
approve the labeling claims that are necessary or desirable for the
successful commercialization of our product candidates.
Clinical
drug development involves a lengthy and expensive process with
uncertain timelines and uncertain outcomes. If clinical studies of
our product candidates are prolonged or delayed, we may be unable
to obtain required regulatory approvals, and therefore be unable to
commercialize our product candidates on a timely basis or at
all.
To obtain the necessary regulatory
approvals to market and sell any of our product candidates, we must
demonstrate through extensive preclinical and clinical studies that
our products are safe and effective in humans. Clinical testing is
expensive and can take many years to complete, and its outcome is
inherently uncertain. Failure can occur at any time during the
clinical study process. The results of preclinical and early
clinical studies of our product candidates may not be predictive of
the results of later-stage clinical studies. For example, the
positive results generated to date in clinical studies for our
product candidates do not ensure that later clinical studies will
demonstrate similar results. Product candidates in later stages of
clinical studies may fail to show the desired safety and efficacy
traits despite having progressed through preclinical studies and
initial clinical studies. A number of companies in the
pharmaceutical or biopharmaceutical industry, including us, have
suffered significant setbacks in advanced clinical studies due to
lack of efficacy or adverse safety profiles, notwithstanding
promising results in earlier studies. Our future clinical study
results may not be successful.
Clinical studies must be conducted
in accordance with the legal requirements, regulations and
guidelines of the FDA, EMA and comparable foreign regulatory
authorities, and are subject to oversight by these governmental
agencies and Institutional Review Boards (IRBs) at the medical
institutions where the clinical studies are conducted. In addition,
clinical studies must be conducted with supplies of our product
candidates produced under cGMP and other requirements. We depend on
medical institutions and CROs to conduct our clinical studies in
compliance with cGCP standards. To the extent the CROs fail to
enroll participants for our clinical studies, fail to conduct the
study to cGCP standards or are delayed for a significant time in
the execution of studies, including achieving full enrollment, we
may be affected by increased costs, program delays or both, which
may harm our business.
To date, neither we nor our
collaboration partners have completed all clinical studies required
for the approval of any of our product candidates.
The completion of clinical studies
for our product candidates may be delayed, suspended or terminated
as a result of many factors, including but not limited to:
|
• |
the delay or refusal of regulators or IRBs to authorize us to
commence or amend a clinical study at a prospective study site or
changes in regulatory requirements, policies and guidelines;
|
|
• |
delays or failure to reach agreement on acceptable terms with
prospective CROs and clinical study sites, the terms of which can
be subject to extensive negotiation and may vary significantly
among different CROs and study sites;
|
|
• |
delays in patient enrollment and variability in the number and
types of patients available for clinical studies;
|
|
• |
the inability to enroll a sufficient number of patients in
studies to ensure adequate statistical power to detect
statistically significant treatment effects;
|
|
• |
negative or inconclusive results, which may require us to
conduct additional preclinical or clinical studies or to abandon
projects that we expected to be promising;
|
|
• |
safety or tolerability concerns, which could cause us to
suspend or terminate a study if we find that the participants are
being exposed to unacceptable health risks;
|
|
• |
regulators or IRBs requiring that we or our investigators
suspend or terminate clinical research for various reasons,
including noncompliance with regulatory requirements or safety
concerns, among others;
|
|
• |
lower than anticipated retention rates of patients and
volunteers in clinical studies;
|
|
• |
our CROs or clinical study sites failing to comply with
regulatory requirements or meet their contractual obligations to us
in a timely manner, or at all, deviating from the protocol or
dropping out of a study;
|
|
• |
delays relating to adding new clinical study sites;
|
|
• |
difficulty in maintaining contact with patients after
treatment, resulting in incomplete data;
|
|
• |
delays in establishing the appropriate dosage levels;
|
|
• |
the quality or stability of the product candidate falling
below acceptable standards;
|
|
• |
the inability to produce or obtain sufficient quantities of
the product candidate to complete clinical studies; and
|
|
• |
exceeding budgeted costs due to difficulty in accurately
predicting costs associated with clinical studies.
|
Any delays in completing our
clinical studies will increase our costs, slow our product
candidate development and approval process, and jeopardize our
ability to commence product sales and generate sales revenues. Any
of these occurrences may significantly harm our business, financial
condition and prospects. In addition, many of the factors that
cause, or lead to, a delay in the commencement or completion of
clinical studies may also ultimately lead to the denial of
regulatory approval of our product candidates.
Even if
we obtain and maintain approval for our product candidates from one
jurisdiction, we may never obtain approval for our product
candidates in other jurisdictions, which would limit our market
opportunities and adversely affect our business.
Sales by us of our approved drugs
will be subject to US and non-US regulatory requirements governing
clinical studies and regulatory approval, and we plan to seek
regulatory approval to commercialize our product candidates in the
US, the European Economic Area (EEA), and other countries. Clinical
studies conducted in one country may not be accepted by regulatory
authorities in other countries, and regulatory approval in one
country does not ensure approval in any other country, while a
failure or delay in obtaining regulatory approval in one country
may have a negative effect on the regulatory approval process in
others. For example, approval in the US by the FDA does not ensure
approval by the regulatory authorities in other countries or
jurisdictions, and similarly, approval by a non-US regulatory
authority, such as the EMA, does not ensure approval by regulatory
authorities in other countries, including by the FDA. Approval
processes and regulatory requirements vary among countries and can
involve additional drug testing and validation and additional
administrative review periods. Even if a drug is approved, the FDA
or EMA, as the case may be, may limit the indications for which the
drug may be marketed, require extensive warnings on the drug
labeling, or require expensive and time-consuming clinical studies
or reporting as conditions of approval. In many countries outside
the US, a product candidate must be approved for reimbursement
before it can be approved for sale in that country. In some cases,
the price that would be charged for a drug is also subject to
approval. Regulatory authorities in other countries also have their
own requirements for approval of product candidates with which we
must comply prior to marketing in those countries. Obtaining non-US
regulatory approvals and compliance with such non-US regulatory
requirements could result in significant delays, difficulties and
costs for us and could delay or prevent the introduction of our
current and any future drugs, in certain countries. If we fail to
comply with regulatory requirements in international markets or to
obtain and maintain required approvals, or if regulatory approvals
in international markets are delayed, our target market will be
reduced and our ability to realize the full market potential of our
product candidates will be unrealized.
Even if
our product candidates obtain regulatory approval, we will be
subject to ongoing obligations and continued regulatory review,
which may result in significant additional expense. Additionally,
our product candidates, if approved, could be subject to labeling
and other restrictions and market withdrawal and we may be subject
to penalties if we fail to comply with regulatory requirements or
experience unanticipated problems with our products.
If a marketing authorization is
obtained for any of our product candidates, the product will remain
subject to continual regulatory review and therefore authorization
could be subsequently withdrawn or restricted. Any regulatory
approvals that we receive for our product candidates may also be
subject to limitations on the approved indicated uses for which the
product may be marketed or to the conditions of approval, or
contain requirements for potentially costly post-marketing testing,
including Phase 4 clinical studies and surveillance to monitor the
safety and efficacy of the product candidate. In addition, if the
FDA or a comparable foreign regulatory authority approves any of
our product candidates, we will be subject to ongoing regulatory
obligations and oversight by regulatory authorities, including with
respect to the manufacturing processes, labeling, packing,
distribution, adverse event reporting, storage, advertising and
marketing restrictions, and record-keeping and, potentially, other
post-marketing obligations, all of which may result in significant
expense and limit our or our collaboration partners’ ability to
commercialize such products. These requirements include submissions
of safety and other post-marketing information and reports,
registration, as well as continued compliance with cGMP and cGCP
requirements for any clinical studies that we conduct
post-approval. Later discovery of previously unknown problems with
a product, including AEs of unanticipated severity or frequency, or
with our third-party manufacturers or manufacturing processes, or
failure to comply with regulatory requirements, may result in,
among other things:
|
• |
restrictions on the marketing or manufacturing of the product,
withdrawal of the product from the market, or voluntary or
mandatory product recalls;
|
|
• |
fines, warning letters or holds on clinical studies;
|
|
• |
refusal by the FDA or an applicable foreign regulatory
authority to approve pending applications or supplements to
approved applications filed by us or our collaborations partners,
or suspension or revocation of product license approvals;
|
|
• |
regulatory constraints in promotion and distribution of drug
products in various markets;
|
|
• |
product seizure or detention, or refusal to permit the import
or export of products; and
|
|
• |
injunctions or the imposition of civil or criminal
penalties.
|
If any of these events occurs, our
ability to sell such product may be impaired, and we may incur
substantial additional expense to comply with regulatory
requirements, which could materially adversely affect our business,
financial condition and results of operations. The FDA’s or those
of an applicable foreign regulatory authority’s policies may change
and additional government regulations may be enacted that could
prevent, limit or delay regulatory approval of our product
candidates. If we are slow or unable to adapt to changes in
existing requirements or the adoption of new requirements or
policies, or if we are not able to maintain regulatory compliance,
we may lose any marketing approval that we may have obtained, which
would adversely affect our business, prospects and ability to
achieve or sustain profitability.
We have
conducted and may in the future conduct clinical studies for our
product candidates outside the US, and the FDA and applicable
foreign regulatory authorities may not accept data from such
studies.
We have conducted and may in the
future choose to conduct one or more of our clinical studies
outside the US, including in Germany, Austria, Denmark, Sweden,
Finland, the UK, Poland, Spain and the Netherlands. The acceptance
of study data from clinical studies conducted outside the US or
another jurisdiction by the FDA or applicable foreign regulatory
authority may be subject to certain conditions. In cases where data
from foreign clinical studies are intended to serve as the basis
for marketing approval in the US, the FDA will not approve the
application on the basis of foreign data alone unless the following
are true: the data are applicable to the US population and US
medical practice; the studies were performed by clinical
investigators of recognized competence; and the data are considered
valid without the need for an on-site inspection by the FDA or, if
the FDA considers such an inspection to be necessary, the FDA is
able to validate the data through an on-site inspection or other
appropriate means. Additionally, the FDA’s clinical study
requirements, including sufficient size of patient populations and
statistical powering, must be met. Many foreign regulatory bodies
have similar requirements. In addition, such foreign studies would
be subject to the applicable local laws of the foreign
jurisdictions in which the studies are conducted. There can be no
assurance that the FDA or any applicable foreign regulatory
authority will accept data from studies conducted outside of the US
or the applicable jurisdiction. If the FDA or any applicable
foreign regulatory authority does not accept such data, it would
result in the need for additional studies, which would be costly
and time-consuming and delay aspects of our business plan, and
which may result in our drugs or product candidates not receiving
approval or clearance for commercialization in the applicable
jurisdiction.
Enacted
and future legislation may increase the difficulty and cost for us
to obtain marketing approval of and commercialize our product
candidates and may affect the prices we may set.
In the US and the EU, there have
been a number of legislative and regulatory changes and proposed
changes regarding the healthcare system. These changes could
prevent or delay marketing approval of our product candidates and
restrict or regulate post-approval activities and affect our
ability to profitably sell any products for which we obtain
marketing approval.
In the US, the Medicare
Prescription Drug, Improvement, and Modernization Act of 2003
(Medicare Modernization Act), changed the way Medicare covers and
pays for pharmaceutical and biopharmaceutical products. The
legislation expanded Medicare coverage for drug purchases by the
elderly and introduced a new reimbursement methodology based on
average sale prices for physician-administered drugs. In addition,
this legislation provided authority for limiting the number of
drugs that will be covered in any therapeutic class. Cost-reduction
initiatives and other provisions of this legislation could decrease
the coverage and price that we receive for any approved products.
Although the Medicare Modernization Act applies only to drug
benefits for Medicare beneficiaries, private payors often follow
Medicare coverage policy and payment limitations in setting their
own reimbursement rates. Therefore, any reduction in reimbursement
that results from the Medicare Modernization Act may result in a
similar reduction in payments from private payors.
In March 2010, former President
Obama signed into law the Patient Protection and Affordable Care
Act, as amended by the Health Care and Education Reconciliation Act
(HCERA) (collectively, the Health Care Reform Law), a sweeping law
intended to broaden access to health insurance, reduce or constrain
the growth of healthcare spending, enhance remedies against fraud
and abuse, add new transparency requirements for health care and
health insurance industries, impose new taxes and fees on the
health industry and impose additional health policy reforms. The
Health Care Reform Law, among other things, increased the rebates a
manufacturer must pay to the Medicaid program; addressed a new
methodology by which rebates owed by manufacturers under the
Medicaid Drug Rebate Program are calculated for drugs that are
inhaled, infused, instilled, implanted or injected; established a
new Medicare Part D coverage gap discount program, in which
manufacturers must provide 50% point-of-sale discounts on products
covered under Part D; and implemented payment system reforms,
including a national pilot program on payment bundling to encourage
hospitals, physicians and other providers to improve the
coordination, quality and efficiency of certain healthcare services
through bundled payment models. Further, the new law imposed a
significant annual fee on companies that manufacture or import
branded prescription drug products. Substantial new provisions
affecting compliance were enacted, which may affect our business
practices with healthcare practitioners. On July 24, 2020 and
September 13, 2020, the Trump administration announced several
executive orders related to prescription drug pricing that attempt
to implement several of the administration’s proposals. As a
result, the FDA released a final rule on September 24, 2020,
effective November 30, 2020, providing guidance for states to build
and submit importation plans for drugs from Canada. Further, on
November 20, 2020, the US Department of Health and Human Services,
or HHS, finalized a regulation removing safe harbor protection for
price reductions from pharmaceutical manufacturers to plan sponsors
under Part D, either directly or through pharmacy benefit managers,
unless the price reduction is required by law. The implementation
of the rule has been delayed by the Biden administration from
January 1, 2022 to January 1, 2023 in response to ongoing
litigation. The rule also creates a new safe harbor for price
reductions reflected at the point-of-sale, as well as a safe harbor
for certain fixed fee arrangements between pharmacy benefit
managers and manufacturers, the implementation of which have also
been delayed until January 1, 2023. On November 20, 2020, the
Centers for Medicare and Medicaid Services (CMS) issued an interim
final rule implementing former President Trump’s Most Favored
Nation executive order, which would tie Medicare Part B payments
for certain physician-administered drugs to the lowest price paid
in other economically advanced countries. The Most Favored Nation
regulations mandate participation by identified Medicare Part B
providers and will apply in all U.S. states and territories for a
seven-year period beginning January 1, 2021 and ending December 31,
2027. On December 28, 2020, the US District Court in Northern
California issued a nationwide preliminary injunction against
implementation of the interim final rule. It is unclear whether the
Biden administration will work to reverse these measures or pursue
similar policy initiatives. At the state level, legislatures have
increasingly passed legislation and implemented regulations
designed to control pharmaceutical and biological product pricing,
including price or patient reimbursement constraints, discounts,
restrictions on certain product access and marketing cost
disclosure and transparency measures, and, in some cases, designed
to encourage importation from other countries and bulk
purchasing.
In 2020, we continued to face
uncertainties because of continued US federal legislative and
administrative efforts to repeal, substantially modify or
invalidate some of the provisions of the Health Care Reform Law. In
January 2017, Congress voted to adopt a budget resolution for the
fiscal year 2017 that authorized the implementation of legislation
that would repeal portions of the Health Care Reform Law. On
December 14, 2018, a federal judge in Texas ruled that the Health
Care Reform Law is unconstitutional in its entirety because the
“individual mandate” was repealed by Congress as part of the 2017
Tax Act. On December 18, 2019, the Fifth Circuit Court of Appeals
upheld the lower court’s decision that the Health Care Reform Law
was unconstitutional. On March 2, 2020, the US Supreme Court
granted certiorari to review the case and oral arguments were held
on November 10, 2020. On January 28, 2021, President Biden issued
an executive order to initiate a special enrollment period from
February 15, 2021 through May 15, 2021 for purposes of obtaining
health insurance coverage through the Health Care Reform Law
marketplace. The executive order also instructs certain
governmental agencies to review and reconsider their existing
policies and rules that limit access to healthcare, including among
others, reexamining Medicaid demonstration projects and waiver
programs that include work requirements, and policies that create
unnecessary barriers to obtaining access to health insurance
coverage through Medicaid or the ACA. On June 17, 2021, the U.S.
Supreme Court rejected the challenge to the Health Care Reform Law
after finding that plaintiffs do not have standing to challenge the
constitutionality of the statute. It is unclear what effect similar
litigation, other efforts to repeal and replace the Health Care
Reform Law and the healthcare reform measures of the Biden
administration will have on the status of the ACA. Litigation and
legislation over the Health Care Reform Law are likely to continue,
with unpredictable and uncertain results. Congress also could
consider subsequent legislation to replace elements of the Health
Care Reform Law that are repealed. There is no assurance that the
Health Care Reform Law, as currently enacted or as amended in the
future, will not adversely affect our business and financial
results, and we cannot predict how future federal or state
legislative or administrative changes relating to healthcare reform
will affect our business.
Moreover, other legislative changes
have also been proposed and adopted in the US since the Health Care
Reform Law was enacted. On August 2, 2011, the Budget Control Act
of 2011, among other things, created measures for spending
reductions by Congress. A Joint Select Committee on Deficit
Reduction, tasked with recommending a targeted deficit reduction of
at least USD 1.2 trillion for the years 2013 through 2021, was
unable to reach the required goals, thereby triggering the
legislation’s automatic reduction to several government programs.
This includes aggregate reductions in Medicare payments to
providers of 2% per fiscal year, which went into effect on April 1,
2013 and, due to subsequent legislative amendments, including the
Infrastructure Investment and Jobs Act, will remain in effect
through 2031 unless additional Congressional action is taken. The
Coronavirus Aid, Relief, and Economic Security Act and subsequent
legislation suspended the 2% Medicare sequester from May 1, 2020
through March 31, 2022. On January 2, 2013, former President Obama
signed into law the American Taxpayer Relief Act of 2012 which,
among other things, further reduced Medicare payments to several
providers, including hospitals, imaging centers and cancer
treatment centers, and increased the statute of limitations period
for the government to recover overpayments to providers from 3 to 5
years. Further, on March 11, 2021, President Biden signed the
American Rescue Plan Act of 2021 into law, which eliminates the
statutory Medicaid drug rebate cap, currently set at 100% of a
drug’s average manufacturer price, for single source and innovator
multiple source drugs, beginning January 1, 2024. The current U.S.
administration continues to focus heavily on drug pricing issues
and Congress has introduced a multitude of legislative proposals
aimed at drug pricing. In addition, Congress is considering
additional health reform measures as part of the budget
reconciliation process. These new laws may result in additional
reductions in Medicare and other healthcare funding, which could
have a material adverse effect on our customers and accordingly,
our financial operations.
Additionally, in the EU, the new
clinical trial regulation is scheduled to come into force on
January 31, 2022. This new legislation will enforce the
centralization of clinical trial applications and approvals, which
will eliminate redundancy, but in some cases, this may extend
timelines for clinical study approvals, due to potentially longer
wait times. Austerity measures in certain European nations may also
affect the prices we are able to seek if our products are approved.
Both in the US and in the EU, legislative and regulatory proposals
have been made to expand post-approval requirements and restrict
sales and promotional activities for pharmaceutical and
biopharmaceutical products. We do not know whether additional
legislative changes will be enacted, whether the regulations,
guidance or interpretations will be changed, or what the impact of
such changes on the marketing approvals of our product candidates,
if any, may be.
We could
be subject to liabilities under environmental, health and safety
laws or regulations, or fines, penalties or other sanctions, if we
fail to comply with such laws or regulations or otherwise incur
costs that could have a material adverse effect on the success of
our business.
We are subject to numerous
environmental, health and safety laws, regulations, and permitting
requirements, including those governing laboratory procedures,
decontamination activities, and the handling, transportation, use,
remediation, storage, treatment and disposal of hazardous
materials, human substances and wastes. Our operations involve the
use of hazardous and flammable materials, including chemicals and
biological materials that produce hazardous waste products. We
generally contract with third parties for the disposal of these
materials and wastes. We cannot eliminate the risk of contamination
or injury from these materials or wastes either at our sites or at
third-party disposal sites. In the event of such contamination or
injury, we could be held liable for any resulting damages, and any
liability could exceed our resources. We also could incur
significant costs associated with civil or criminal fines and
penalties. Although we maintain workers’ compensation insurance to
cover us for costs and expenses we may incur due to injuries to our
employees resulting from the use of hazardous materials, human
substances or other work-related injuries, this insurance may not
provide adequate coverage against potential liabilities.
In addition, we may incur
substantial costs in order to comply with current or future
environmental, health and safety laws, regulations or permitting
requirements. Such laws, regulations and requirements are becoming
increasingly more stringent and may impair our research,
development or production efforts. Failure to comply with these
laws, regulations and permitting requirements also may result in
substantial fines, penalties or other sanctions.
Our
relationships with clinical centers are, and potentials customers
and payors will be, subject to applicable anti-kickback, fraud and
abuse and other healthcare laws and regulations, which, if
violated, could expose us to criminal sanctions, civil penalties,
exclusion from government healthcare programs, contractual damages,
reputational harm and diminished profits and future earnings.
Healthcare providers, physicians
and others play a primary role in the recommendation and
prescription of any products for which we obtain marketing
approval. Our future arrangements with third-party payors and
customers may expose us to broadly applicable fraud and abuse and
other healthcare laws and regulations, which constrain the business
or financial arrangements and relationships through which we
market, sell and distribute our products for which we obtain
marketing approval. Restrictions under applicable healthcare laws
and regulations include the following:
|
• |
the US healthcare Anti-Kickback Statute prohibits, among other
things, persons from knowingly and willfully soliciting, offering,
receiving or providing remuneration, directly or indirectly, in
cash or in kind, to induce or reward either the referral of an
individual for, or the purchase, order or recommendation of, any
good or service, for which payment may be made under US government
healthcare programs such as Medicare and Medicaid;
|
|
• |
the US False Claims Act imposes criminal and civil penalties,
including civil whistleblower or qui tam actions, against individuals or
entities for knowingly presenting, or causing to be presented, to
the US government, claims for payment that are false or fraudulent
or making a false statement to avoid, decrease or conceal an
obligation to pay money to the federal government;
|
|
• |
the US HIPAA imposes criminal and civil liability for
executing a scheme to defraud any healthcare benefit program or
making false statements relating to healthcare matters;
|
|
• |
the HIPAA, as amended by the Health Information Technology for
Economic and Clinical Health Act, imposes obligations, including
mandatory contractual terms, with respect to safeguarding the
privacy, security and transmission of individually identifiable
health information;
|
|
• |
the transparency requirements under the Health Care Reform Law
require manufacturers of drugs, devices, biologics and medical
supplies to report to the US Department of Health and Human
Services information related to payments and other transfers of
value made by such manufacturers to physicians and teaching
hospitals, and ownership and investment interests held by
physicians or their immediate family members; and
|
|
• |
analogous laws and regulations, such as state anti-kickback
and false claims laws, will apply to sales or marketing
arrangements, consultancy and service agreements, and claims
involving healthcare items or services reimbursed by
nongovernmental third-party payors, including private insurers, and
some state laws require pharmaceutical and biopharmaceutical
companies to comply with the pharmaceutical and biopharmaceutical
industries’ voluntary compliance guidelines and the relevant
compliance guidance promulgated by the federal government, in
addition to requiring manufacturers to report information related
to payments to physicians and other healthcare providers or
marketing expenditures.
|
Because of the breadth of these
laws and the narrowness of the statutory exceptions and safe
harbors available under the US federal Anti-Kickback Statute, it is
possible that some of our future business activities could be
subject to challenge under one or more of such laws. In addition,
recent healthcare-reform legislation has strengthened these laws.
For example, the Health Care Reform Law, among other things, amends
the intent requirement of the federal anti-kickback and criminal
healthcare fraud statutes. A person or entity no longer needs to
have actual knowledge of this statute or specific intent to violate
it. Moreover, the Health Care Reform Law provides that the
government may assert that a claim including items or services
resulting from a violation of the federal anti-kickback statute
constitutes a false or fraudulent claim for purposes of the False
Claims Act.
Jurisdictions outside of the US
have enacted laws and regulations defining the framework of
business practices of pharmaceutical organizations in their
interactions with government offices, medical institutions and
healthcare professionals (HCP) in order to safeguard the
independence of medical judgement and of prescription and
purchasing decisions. These regulations typically prohibit
illegitimate payments and other transfers of values to
institutional players and HCPs and regulate the bases for their
remuneration, such as for consultancy and other service
arrangements, as well as the reimbursement of costs; in certain
jurisdictions, regulations prescribe the disclosure of the existing
relationships and/or the remunerations paid. In addition to
government regulations, pharmaceutical industry associations, such
as the European Federation of Pharmaceutical Industries and
Associations (EFPIA), of which we have been a member since 2021,
have enacted industry codes of conduct providing their own rules of
compliance for their members’ interactions with government offices,
medical institutions and HCPs.
Efforts to ensure that our business
arrangements with third parties will comply with applicable
healthcare laws and regulations will involve substantial costs. It
is possible that governmental authorities will conclude that our
business practices may not comply with current or future statutes,
regulations or case law involving applicable fraud and abuse or
other healthcare laws and regulations. If our operations are found
to be in violation of any of these laws or any other governmental
regulations that may apply to us, we may be subject to significant
civil, criminal and administrative penalties, damages, fines,
exclusion from US government-funded healthcare programs, such as
Medicare and Medicaid, other foreign healthcare reimbursement and
procurement programs, and the curtailment or restructuring of our
operations. If any of the physicians or other providers or entities
with whom we expect to do business with is found to be not in
compliance with applicable laws, they may be subject to criminal,
civil or administrative sanctions, including exclusions from
government-funded healthcare programs.
Risks
from the improper conduct of employees, agents, contractors, or
collaborators could adversely affect our reputation and our
business, prospects, operating results, and financial
condition.
We cannot ensure that our
compliance controls, policies, and procedures will in every
instance protect us from acts committed by our employees, agents,
contractors, or collaborators, which would violate the laws or
regulations of the jurisdictions in which we operate, including,
without limitation, healthcare, employment, foreign corrupt
practices, environmental, competition, and patient privacy and
other privacy laws and regulations. Such improper actions could
subject us to civil or criminal investigations, and monetary and
injunctive penalties, and could adversely impact our operating
results, our ability to conduct business and our reputation.
We are exposed to the risk of
employee fraud or other misconduct. Misconduct by employees could
include intentional failures to comply with FDA or EMA regulations,
to provide accurate information to the FDA or the EMA, or
intentional failures to report financial information or data
accurately or to disclose unauthorized activities to us. Employee
misconduct could also involve the improper use of information
obtained in the course of clinical studies, which could result in
regulatory sanctions and serious harm to our reputation. In April
2021 we amended our code of conduct, but it is not always possible
to identify and deter employee misconduct, and the precautions we
take to detect and prevent this activity may not be effective in
controlling unknown or unmanaged risks or losses or in protecting
us from governmental investigations or other actions or lawsuits
stemming from a failure to comply with these laws or regulations.
If any such actions are instituted against us, and we are not
successful in defending ourselves or asserting our rights, those
actions could have a significant impact on our business, including
the imposition of significant fines or other sanctions.
Our
business activities may be subject to the Foreign Corrupt Practices
Act (FCPA), and similar anti-bribery and anti-corruption
laws.
Our business activities may be
subject to the FCPA and similar anti-bribery or anti-corruption
laws, regulations or rules of other countries in which we operate,
including the UK Bribery Act. The FCPA generally prohibits
offering, promising, giving or authorizing others to give anything
of value, either directly or indirectly, to a non-US government
official in order to influence official action, or otherwise obtain
or retain business. The FCPA also requires public companies to make
and keep books and records that accurately and fairly reflect the
transactions of the corporation, and to devise and maintain an
adequate system of internal accounting controls. Our business is
heavily regulated and therefore involves significant interaction
with public officials, including officials of non-US governments.
Additionally, in many other countries, the healthcare providers who
prescribe pharmaceuticals or biopharmaceuticals and the
investigators who perform our studies are employed by their
government, and the purchasers of pharmaceuticals are government
entities; therefore, our dealings with these prescribers and
purchasers are subject to regulation under the FCPA. The Securities
and Exchange Commission (SEC) and the Department of Justice have
increased their FCPA enforcement activities with respect to
pharmaceutical companies. There is no certainty that all of our
employees, agents, contractors or collaborators, or those of our
affiliates, will comply with all applicable laws and regulations,
particularly given the high level of complexity of these laws.
Violations of these laws and regulations could result in fines,
criminal sanctions against us, our officers or our employees, the
closing down of our facilities, requirements to obtain export
licenses, cessation of business activities in sanctioned countries,
implementation of compliance programs, and prohibitions on the
conduct of our business. Any such violations could include
prohibitions on our ability to offer our products in one or more
countries and could materially damage our reputation, our brand,
our international expansion efforts, our ability to attract and
retain employees, and our business, prospects, operating results
and financial condition.
Risks related to our common
shares
The
price of our common shares may be volatile and may fluctuate due to
factors beyond our control.
The share prices of publicly traded
emerging pharmaceutical, biopharmaceutical and drug discovery and
development companies have been highly volatile and are likely to
remain highly volatile in the future. The market price of our
common shares may fluctuate significantly due to a variety of
factors, including:
|
• |
positive or negative results of testing and clinical studies
by us, strategic partners, or competitors;
|
|
• |
delays in entering into strategic relationships with respect
to development and/or commercialization of our product candidates
or entry into strategic relationships on terms that are not deemed
to be favorable to us;
|
|
• |
the sentiment of retail investors, including the perception of
our clinical trial results by such retail investors, which
investors may be subject to the influence of information provided
by social media, third party investor websites and independent
authors distributing information on the internet;
|
|
• |
technological innovations or commercial product introductions
by us or our collaboration partners or competitors;
|
|
• |
changes in government regulations;
|
|
• |
developments concerning proprietary rights, including patents
and litigation matters;
|
|
• |
public concern relating to the commercial value or safety of
any of our product candidates;
|
|
• |
financing or other corporate transactions;
|
|
• |
publication of research reports or comments by securities or
industry analysts or key opinion leaders;
|
|
• |
general market conditions in the pharmaceutical or
biopharmaceutical industry or in the economy as a whole; or
|
|
• |
other events and factors beyond our control.
|
Broad market and industry factors
may materially affect the market price of companies’ stock,
including ours, regardless of actual operating performance.
Furthermore, issuers such as ourselves, whose securities have
historically had limited trading volumes and/or have been
susceptible to relatively high volatility levels, can be
particularly vulnerable to short-seller attacks and trading in our
common shares by non-fundamental investors such as hedge funds and
others who may enter and exit positions in our common shares
frequently and suddenly, causing increased volatility of our share
price. Short selling is the practice of selling securities that the
seller does not own but rather has borrowed or intends to borrow
from a third party with the intention of buying identical
securities at a later date to return to the lender, and profit from
a decline in the value of the securities in the process. The
publication of any commentary by short sellers with the intent of
creating negative market momentum may bring about a temporary, or
possibly long-term, decline in the market price of our common
shares.
There
is only a limited free float of our common shares; this may have a
negative impact on the liquidity of and the market price for our
common shares.
As of the date hereof, certain
principal shareholders controlling 5% or more of our common shares
as well as our executive officers and directors together
beneficially own approximately 61.2% of our common shares. The
limited free float may have a negative impact on the liquidity of
our common shares and result in a low trading volume of our common
shares, which could adversely affect the price of our common
shares.
Certain of our existing shareholders exercise significant control
over us, and your interests may conflict with the interests of such
shareholders.
Certain principal shareholders as
well as our executive officers and directors together beneficially
own approximately 61.2% of our common shares. Depending on the
level of attendance at our general meetings of shareholders, these
shareholders may be in a position to determine the outcome of
decisions taken at any such general meeting. To the extent that the
interests of these shareholders may differ from the interests of
the Company’s other shareholders, the latter may be disadvantaged
by any action that these shareholders may seek to pursue. Among
other consequences, this concentration of ownership may have the
effect of delaying or preventing a change in control and might
therefore negatively affect the market price of our common
shares.
Future sales, or the possibility of future sales, of a substantial
number of our common shares could adversely affect the price of our
common shares.
Future sales of a substantial
number of our common shares, or the perception that such sales will
occur, could cause a decline in the market price of our common
shares. If certain of our shareholders sell substantial amounts of
common shares in the public market, or the market perceives that
such sales may occur, the market price of our common shares and our
ability to raise capital through an issue of equity securities in
the future could be adversely affected. We also entered into a
registration rights agreement in connection with the Series E
Private Placement with certain investors in the Series E Private
Placement, pursuant to which we agreed under certain circumstances
to file a registration statement to register the resale of the
common shares held by certain of our existing shareholders, as well
as to cooperate in certain public offerings of such common shares.
In October 2020 and August 2018, we filed registration statements
on Form F-3 to register the resale of two of our shareholder’s
common shares pursuant to the requirements of their registration
rights agreements. In addition, in 2019, we adopted a new omnibus
equity incentive plan under which we have the discretion to grant a
broad range of equity-based awards to eligible participants. These
shares were registered pursuant to the registration statement on
Form S-8 that we filed with the SEC and, therefore, can be freely
sold in the public market upon issuance, subject to volume
limitations applicable to affiliates. If a large number of our
common shares are sold in the public market after they become
eligible for sale, the sales could reduce the trading price of our
common shares and impede our ability to raise future capital.
We
have broad discretion in the use of our cash and cash equivalents
and short-term financial assets (liquidity) and may not use them
effectively.
Our management has broad discretion
in the application of our cash and cash equivalents and short-term
financial assets. Our or our collaboration partners’ decisions
concerning the allocation of research, development, collaboration,
management and financial resources toward particular product
candidates or therapeutic areas may not lead to the development of
any viable commercial product and may divert resources away from
better opportunities. If we make incorrect determinations regarding
the viability or market potential of any of our programs or product
candidates or misread trends in the pharmaceutical or
biopharmaceutical industry, in particular for neurodegenerative
diseases, our business, financial condition and results of
operations could be materially adversely affected. As a result, we
may fail to capitalize on viable commercial products or profitable
market opportunities, be required to forego or delay pursuit of
opportunities with other product candidates or other diseases and
disease pathways that may later prove to have greater commercial
potential than those we choose to pursue, or relinquish valuable
rights to such product candidates through collaboration, licensing
or other royalty arrangements in cases in which it would have been
advantageous for us to invest additional resources to retain sole
development and commercialization rights.
We do
not expect to pay dividends in the foreseeable future.
We have not paid any dividends
since our incorporation. Even if future operations lead to
significant levels of distributable profits, we currently intend
that any earnings will be reinvested in our business and that
dividends will not be paid until we have an established revenue
stream to support continuing dividends. Based on Swiss law and our
articles of association, the declaration of dividends requires a
resolution passed by a simple majority of the votes cast at a
shareholders’ meeting regardless of abstentions and empty or
invalid votes. The proposal to pay future dividends to shareholders
will in addition effectively be at the discretion of our board of
directors after considering various factors including our business
prospects, liquidity requirements, financial performance and new
product development. In addition, payment of future dividends is
subject to certain limitations pursuant to Swiss law or by our
articles of association compliance with which must be confirmed by
our auditors. Accordingly, investors cannot rely on dividend income
from our common shares and any returns on an investment in our
common shares will likely depend entirely upon any future
appreciation in the price of our common shares.
We
are a Swiss corporation. The rights of our shareholders may be
different from the rights of shareholders in companies governed by
the laws of US jurisdictions.
We are a Swiss corporation. Our
corporate affairs are governed by our articles of association and
by the laws governing companies, including listed companies,
incorporated in Switzerland. The rights of our shareholders and the
responsibilities of members of our board of directors may be
different from the rights and obligations of shareholders and
directors of companies governed by the laws of US jurisdictions. In
the performance of its duties, our board of directors is required
by Swiss law to consider the interests of our Company first, then
of our shareholders, our employees and other stakeholders, in all
cases, with due observation of their fiduciary duties of care and
loyalty. It is possible that some of these parties will have
interests that are different from, or in addition to, your
interests as a shareholder. Swiss corporate law limits the ability
of our shareholders to challenge resolutions made or other actions
taken by our board of directors in court. Our shareholders
generally are not permitted to file a suit to reverse a decision or
an action taken by our board of directors but are instead only
permitted to seek damages for breaches of their fiduciary duties by
the directors. As a matter of Swiss law, shareholder claims against
a member of our board of directors for breach of fiduciary duty
would have to be brought in Lausanne, Switzerland, or the country
in which the relevant member of our board of directors is
domiciled. In addition, under Swiss law, any claims by our
shareholders against us must be in principle brought exclusively in
Lausanne, Switzerland (except for certain US securities and other
claims that may be brought in US federal court).
Our
common shares are issued under the laws of Switzerland, which may
not protect investors in a similar fashion afforded by
incorporation in a US state.
We are organized under the laws of
Switzerland. There can be no assurance that Swiss law will not
change in the future in a way detrimental to shareholders or that
it will serve to protect investors in a similar fashion afforded
under corporate law principles in the US, which could adversely
affect the rights of investors.
Our
status as a Swiss corporation may limit our flexibility with
respect to certain aspects of capital management and may cause us
to be unable to make distributions without subjecting our
shareholders to Swiss withholding tax.
Swiss law allows our shareholders
to authorize share capital that can be issued by the board of
directors without additional shareholder approval. This
authorization is limited to 50% of the existing registered share
capital and must be renewed by the shareholders every 2 years.
Additionally, as a principle, Swiss law grants pre-emptive
subscription rights to existing shareholders to subscribe to any
new issuance of shares. Any common share capital increase
resolution preserving pre-emptive subscription rights expires after
3 months and requires a simple majority of the votes cast at the
shareholder’s meeting regardless of abstentions and empty or
invalid votes. Swiss law also does not provide as much flexibility
in the various terms that can attach to different classes of shares
as do the laws of some other jurisdictions. Swiss law also reserves
for approval by shareholders certain corporate actions over which a
board of directors would have authority in some other
jurisdictions. For example, dividends must be approved by
shareholders. These Swiss law requirements relating to our capital
management may limit our flexibility, and situations may arise in
which greater flexibility would have provided substantial benefits
to our shareholders.
Under Swiss law, a Swiss
corporation may pay dividends only if the corporation has
sufficient distributable profits from previous fiscal years, or if
the corporation has distributable reserves, each as evidenced by
its audited statutory balance sheet. Freely distributable reserves
are generally booked either as “free reserves” or as “capital
contributions” (apports de
capital, contributions received from shareholders) in the
“reserve from capital contributions.” Distributions may be made out
of issued share capital—the aggregate nominal value of a company’s
issued shares—only by way of a capital reduction. As of December
31, 2021, the Company has CHF 432.6 million of reserves from
capital contributions and CHF 1,792,702 of issued share capital
(consisting of 89,635,115 common shares each with a nominal value
of CHF 0.02 and no preferred shares) on its audited statutory
balance sheet. Of the total issued shares and issued share capital,
the Company holds 6,221,617 fully paid-in treasury shares
representing CHF 124,432 of issued share capital.
We expect the aggregate of these
amounts (i.e. reserves from capital contributions and share
capital, less the total losses brought forward, less the treasury
shares, less the lowest legally possible issued share capital and
legal reserve of together CHF 150,000) to represent the potential
amount available for future dividends or capital reductions on a
Swiss withholding tax-free basis. We will not be able to pay
dividends or make other distributions to shareholders on a Swiss
withholding tax-free basis in excess of that amount unless the
Company increases its share capital or its reserves from capital
contributions. We would also be able to pay dividends out of
distributable profits or freely distributable reserves but such
dividends would be subject to Swiss withholding taxes. There can be
no assurance that we will have sufficient distributable profits,
free reserves, reserves from capital contributions or registered
share capital to pay a dividend or effect a capital reduction, that
our shareholders will approve dividends or capital reductions
proposed by us, or that we will be able to meet the other legal
requirements for dividend payments or distributions as a result of
capital reductions.
Generally, Swiss withholding tax of
35% is due on dividends and similar distributions to our
shareholders, regardless of the place of residency of the
shareholder, unless the distribution is made to shareholders out of
(i) a reduction of nominal value or (ii) assuming certain
conditions are met, reserves from capital contributions accumulated
on or after January 1, 1997. A US Holder who qualifies for benefits
under the Convention Between the United States of America and the
Swiss Confederation for the Avoidance of Double Taxation with
Respect to Taxes on Income, which we refer to as the “US-Swiss
Treaty,” may apply for a refund of the tax withheld in excess of
the 15% treaty rate (or in excess of the 5% reduced treaty rate for
qualifying corporate shareholders with at least 10% participation
in our voting stock, or for a full refund in the case of qualified
pension funds). There can be no assurance that we will have
sufficient reserves from capital contributions to pay dividends
free from Swiss withholding tax, or that Swiss withholding tax
rules will not be changed in the future. In addition, we cannot
provide assurance that the current Swiss law with respect to
distributions out of reserves from capital contributions will not
be changed or that a change in Swiss law will not adversely affect
us or our shareholders, in particular as a result of distributions
out of reserves from capital contributions becoming subject to
additional corporate law or other restrictions. In addition, over
the long term, the amount of nominal value available to us for
nominal value reductions or reserves from capital contributions
available to us to pay out as distributions is limited. If we are
unable to make a distribution through a reduction in nominal value
or out of reserves from capital contributions, we may not be able
to make distributions without subjecting our shareholders to Swiss
withholding taxes.
US
shareholders may not be able to obtain judgments or enforce civil
liabilities against us or our executive officers or members of our
board of directors.
We are organized under the laws of
Switzerland and our registered office and domicile is located in
Ecublens, near Lausanne, Canton of Vaud, Switzerland. Moreover, a
number of our directors and executive officers are not residents of
the US, and all or a substantial portion of the assets of such
persons are located outside the US. As a result, it may not be
possible for investors to effect service of process within the US
upon us or upon such persons or to enforce against them judgments
obtained in US courts, including judgments in actions predicated
upon the civil liability provisions of the federal securities laws
of the US. We have been advised by our Swiss counsel that there is
doubt as to the enforceability in Switzerland of original actions,
or of actions for enforcement of judgments of US courts, for civil
liabilities to the extent solely predicated upon the federal and
state securities laws of the US. Original actions against persons
in Switzerland based solely upon the US federal or state securities
laws are governed, among other things, by the principles set forth
in the Swiss Federal Act on Private International Law. This statute
provides that the application of provisions of non-Swiss law by the
courts in Switzerland shall be precluded if the result is
incompatible with Swiss public policy. Additionally, certain
mandatory provisions of Swiss law may be applicable regardless of
any other law that would otherwise apply.
Switzerland and the US do not have
a treaty providing for reciprocal recognition and enforcement of
judgments in civil and commercial matters. The recognition and
enforcement of a judgment of the courts of the US in Switzerland is
governed by the principles set forth in the Swiss Federal Act on
Private International Law. This statute provides in principle that
a judgment rendered by a non-Swiss court may be enforced in
Switzerland only if:
|
• |
the non-Swiss court had jurisdiction pursuant to the Swiss
Federal Act on Private International Law;
|
|
• |
the judgment of such non-Swiss court has become final and
non-appealable;
|
|
• |
the judgment does not contravene Swiss public policy;
|
|
• |
the court procedures and the service of documents leading to
the judgment were in accordance with the due process of law;
and
|
|
• |
no proceeding involving the same parties and the same subject
matter was first brought in Switzerland, or adjudicated in
Switzerland, or was earlier adjudicated in a third state for which
the decision is recognizable in Switzerland.
|
Our
status as a Swiss corporation means that our shareholders enjoy
certain rights that may limit our flexibility to raise capital,
issue dividends and otherwise manage ongoing capital needs.
Swiss law reserves for approval by
shareholders certain corporate actions over which a board of
directors would have authority in some other jurisdictions. For
example, the payment of dividends and cancellation of treasury
shares must be approved by shareholders. Swiss law also requires
that our shareholders themselves resolve, or authorize our board of
directors, to increase our share capital. Although our shareholders
may authorize share capital that can be issued by our board of
directors without additional shareholder approval, Swiss law limits
this authorization to 50% of the issued share capital at the time
of the authorization. The authorization, furthermore, has a limited
duration of up to 2 years and must be renewed by the shareholders
from time to time thereafter in order to be available for raising
capital. Additionally, subject to specified exceptions, including
exceptions explicitly described in our articles of association,
Swiss law grants pre-emptive subscription rights to existing
shareholders to subscribe for new issuances of shares. Swiss law
also does not provide as much flexibility in the various rights and
regulations that can attach to different categories of shares as do
the laws of some other jurisdictions. These Swiss law requirements
relating to our capital management may limit our flexibility, and
situations may arise where greater flexibility would have provided
benefits to our shareholders.
Swiss
law restricts our ability to pay dividends.
See “Item 10. Additional
information—E. Taxation—Swiss tax considerations” for a summary of
certain Swiss tax consequences regarding dividends distributed to
holders of our common shares.
Shareholders in countries with a currency other than Swiss Francs
face additional investment risks from currency exchange rate
fluctuations in connection with their holding of our common
shares
Any future payments of dividends,
if any, will likely be denominated in Swiss Francs. The foreign
currency equivalent of any dividend, if any, paid on our common
shares or received in connection with any sale of our common shares
could be adversely affected by the depreciation of the Swiss Franc
against such other currency.
We
are a foreign private issuer and, as a result, we are not subject
to US proxy rules and are subject to Exchange Act reporting
obligations that, to some extent, are more lenient and less
frequent than those of a US domestic public company.
We are reporting under the Exchange
Act as a non-US company with foreign private issuer status. Because
we qualify as a foreign private issuer under the Exchange Act and
although we are subject to Swiss laws and regulations with regard
to such matters and intend to furnish quarterly financial
information to the SEC, we are exempt from certain provisions of
the Exchange Act that are applicable to US domestic public
companies, including (i) the sections of the Exchange Act
regulating the solicitation of proxies, consents or authorizations
in respect of a security registered under the Exchange Act; (ii)
the sections of the Exchange Act requiring insiders to file public
reports of their stock ownership and trading activities and their
liability for insiders who profit from trades made in a short
period of time; and (iii) the rules under the Exchange Act
requiring the filing with the SEC of quarterly reports on Form 10-Q
containing unaudited financial and other specified information, or
of current reports on Form 8-K, upon the occurrence of specified
significant events. In addition, foreign private issuers are not
required to file their annual report on Form 20-F until 4 months
after the end of each financial year, whereas US domestic issuers
that are accelerated filers are required to file their annual
report on Form 10-K within 75 days after the end of each fiscal
year. Foreign private issuers are also exempt from the Regulation
Fair Disclosure, aimed at preventing issuers from making selective
disclosures of material information. As a result of the above, you
may not have the same protections afforded to shareholders of
companies that are not foreign private issuers.
As a
foreign private issuer and as permitted by the listing requirements
of Nasdaq, we rely on certain home country governance practices
rather than the corporate governance requirements of Nasdaq.
We are a foreign private issuer. As
a result, in accordance with Nasdaq Listing Rule 5615(a)(3), we
comply with home country (in this case, Swiss) governance
requirements and certain exemptions thereunder rather than
complying with certain of the corporate governance requirements of
Nasdaq. Swiss law does not require that a majority of our board of
directors consist of independent directors. Our board of directors
therefore may include fewer independent directors than would be
required if we were subject to Nasdaq Listing Rule 5605(b)(1). In
addition, we are not subject to Nasdaq Listing Rule 5605(b)(2),
which requires that independent directors regularly have scheduled
meetings at which only independent directors are present.
While Swiss law also requires that
our board of directors elects an audit and finance committee from
among its members, as a foreign private issuer, the independence of
the members of such committee is determined by home country
regulations and the conditions of Section 10B of the Securities
Exchange Act, excluding any Nasdaq Listing Rules. Swiss law also
requires that we elect a compensation committee, we follow home
country requirements with respect to such committee and our
compensation, nomination and corporate governance committee is
tasked with certain director nomination and governance
responsibilities as described under “Item 6. Directors, senior
management and employees.” As a result, our practice varies from
the requirements of Nasdaq Listing Rule 5605(d), which sets forth
certain requirements as to the responsibilities, composition and
independence of compensation committees, and from the independent
director oversight of director nominations requirements of Nasdaq
Listing Rule 5605(e).
Furthermore, in accordance with
Swiss law and generally accepted business practices, our articles
of association do not provide quorum requirements generally
applicable to general meetings of shareholders. Our practice thus
varies from the requirement of Nasdaq Listing Rule 5620(c), which
requires an issuer to provide in its bylaws for a generally
applicable quorum, and that such quorum may not be less than
one-third of the outstanding voting stock. Our articles of
association provide for an independent proxy holder elected by our
shareholders, who may represent our shareholders at a general
meeting of shareholders, and we must provide shareholders with an
agenda and other relevant documents for the general meeting of
shareholders. Our practice varies from the requirement of Nasdaq
Listing Rule 5620(b), which sets forth certain requirements
regarding the solicitation of proxies. In addition, we have opted
out of shareholder approval requirements for the issuance of
securities in connection with certain events such as the
acquisition of stock or assets of another company, the
establishment of or amendments to equity-based compensation plans
for employees, a change of control of us, and certain private
placements. To this extent, our practice varies from the
requirements of Nasdaq Listing Rule 5635, which generally requires
an issuer to obtain shareholder approval for the issuance of
securities in connection with such events.
For an overview of our corporate
governance principles, see “Item 16G. Corporate governance.” As a
result of the above, you may not have the same protections afforded
to shareholders of companies that are not foreign private
issuers.
We
may lose our foreign private issuer status, which would then
require us to comply with the Exchange Act’s domestic reporting
regime and cause us to incur significant legal, accounting and
other expenses.
We are a foreign private issuer and
therefore we are not required to comply with all of the periodic
disclosure and current reporting requirements of the Exchange Act
applicable to US domestic issuers. We may no longer be a foreign
private issuer as of June 30, 2022 (or the end of our second fiscal
quarter in any subsequent fiscal year), which would require us to
comply with all of the periodic disclosure and current reporting
requirements of the Exchange Act applicable to US domestic issuers
as of January 1, 2023 (or the first day of the fiscal year
immediately succeeding the end of such second quarter). In order to
maintain our current status as a foreign private issuer, either (a)
a majority of our common shares must be either directly or
indirectly owned of record by non-residents of the US or (b) (i) a
majority of our executive officers or directors may not be US
citizens or residents, (ii) more than 50 percent of our assets
cannot be located in the US and (iii) our business must be
administered principally outside the US. If we lost this status, we
would be required to comply with the Exchange Act reporting and
other requirements applicable to US domestic issuers, which are
more detailed and extensive than the requirements for foreign
private issuers. We may also be required to make changes in our
corporate governance practices in accordance with various SEC and
stock exchange rules. The regulatory and compliance costs to us
under US securities laws if we are required to comply with the
reporting requirements applicable to a US domestic issuer may be
significantly higher than the cost we would incur as a foreign
private issuer. As a result, we expect that a loss of foreign
private issuer status would increase our legal and financial
compliance costs and would make some activities highly
time-consuming and costly. We also expect that if we were required
to comply with the rules and regulations applicable to US domestic
issuers, it would make it more difficult and expensive for us to
obtain director and officer liability insurance, and we may be
required to accept reduced coverage or incur substantially higher
costs to obtain coverage. These rules and regulations could also
make it more difficult for us to attract and retain qualified
members of our board of directors.
If we fail to maintain an effective system of internal control over
financial reporting, we may not be able to accurately report our
financial results or prevent fraud. As a result, shareholders could
lose confidence in our financial and other public reporting, which
would harm our business and the trading price of our common
shares.
Effective
internal controls over financial reporting are necessary for us to
provide reliable financial reports and, together with adequate
disclosure controls and procedures, are designed to prevent fraud,
among other objectives. Any failure to implement required new or
improved controls, or difficulties encountered in their
implementation could cause us to fail to meet our reporting
obligations. In addition, any testing by us conducted in connection
with Section 404 of the Sarbanes-Oxley Act of 2002, or any
subsequent testing by our independent registered public accounting
firm, may reveal deficiencies in our internal controls over
financial reporting, which are deemed to be material weaknesses or
that may require prospective or retroactive changes to our
consolidated financial statements or identify other areas for
further attention or improvement.
Our management
is required to assess the effectiveness of our internal controls
and procedures annually. We ceased to be an emerging growth company
on December 31, 2021, and, as such, we will no longer be able to
avail ourselves of exemptions from various reporting requirements
applicable to other public companies but not to “emerging growth
companies.” For example, Section 404 requires us to perform system
and process evaluation and testing of our internal control over
financial reporting to allow management to report on, and our
independent registered public accounting firm potentially to attest
to, the effectiveness of our internal control over financial
reporting. We previously availed ourselves of the exemption from
the requirement that our independent registered public accounting
firm attest to the effectiveness of our internal control over
financial reporting under Section 404. However, we are no longer
able to avail ourselves of this exemption. Our management is
required to issue an annual report on internal control over
financial reporting, and our independent registered public
accounting firm is now required to undertake an assessment of our
internal control over financial reporting, which could detect
problems that our management’s assessment might not. Undetected
material weaknesses in our internal controls could lead to
financial statement restatements and require us to incur the
expense of remediation. The rules governing the standards that must
be met for our management to assess our internal control over
financial reporting pursuant to Section 404 of the Sarbanes-Oxley
Act are complex and require significant documentation, testing and
possible remediation. These stringent standards require that our
audit committee be advised and regularly updated on management’s
review of internal control over financial reporting in connection
with issuing our consolidated financial statements as of and for
the year ended December 31, 2021.
Moreover, if
we are not able to comply with the requirements of Section 404
applicable to us in a timely manner, or if we or our independent
registered public accounting firm identifies deficiencies in our
internal control over financial reporting that are deemed to be
material weaknesses, the market price of our common shares could
decline, and we could be subject to sanctions or investigations by
the SEC or other regulatory authorities, which would require
additional financial and management resources. Furthermore,
investor perceptions of our company may suffer if deficiencies are
found, and this could cause a decline in the market price of our
common shares. Irrespective of compliance with Section 404, any
failure of our internal control over financial reporting could have
a material adverse effect on our stated operating results and harm
our reputation. If we are unable to implement these requirements
effectively or efficiently, it could harm our operations, financial
reporting, or financial results and could result in an adverse
opinion on our internal control over financial reporting from our
independent registered public accounting firm.
If
securities or industry analysts do not publish research, or publish
inaccurate or unfavorable research, about our business, the price
of our common shares and our trading volume could decline.
The trading market for our common
shares will depend in part on the research and reports that
securities or industry analysts publish about us or our business.
If no or too few securities or industry analysts cover our company,
the trading price for our common shares would likely be negatively
affected. In addition, if one or more of the analysts who cover us
downgrade our common shares or publish inaccurate or unfavorable
research about our business, the price of our common shares would
likely decline. If one or more of these analysts cease coverage of
our company or fail to publish reports on us regularly, demand for
our common shares could decrease, which might cause the price of
our common shares and trading volume to decline.
It is
likely that we were a PFIC for 2019 and 2020. Although we believe
we were not a PFIC for 2021, there can be no assurance that the
Internal Revenue Service will agree. We cannot express any
expectation regarding our PFIC status for 2022 or any future
taxable year. If we are a PFIC for any taxable year during which a
US investor owns our common shares, the investor generally will be
subject to adverse US federal income tax consequences.
Under the Internal Revenue Code of
1986, as amended (the “Code”), we will be a PFIC for any taxable
year in which, after the application of certain look-through rules
with respect to subsidiaries, either (i) 75% or more of our gross
income consists of passive income (the “income test”) or (ii) 50%
or more of the average value of our assets (generally determined on
a quarterly basis) consists of assets that produce, or are held for
the production of, passive income (the “asset test”). Passive
income generally includes dividends, interest, certain non-active
rents and royalties, and gains from financial investments.
Although the application of the
income test to a company like us (whose overall losses from
research and development activities significantly exceed its gross
income) is not entirely clear, we will be a PFIC for any taxable
year under the income test if 75% or more of our gross income (as
determined for U.S. federal income tax purposes) for such year
consists of interest and other passive income. Prior to the
commercialization and sales of any of our product candidates, our
gross income may consist primarily of upfront or milestone payments
(which we believe are active income), grants (which are likely to
be treated as active income) and interest (which is passive
income). The receipt of upfront payments is non-recurring in
nature, and the receipt of grants or milestone payments is subject
to various conditions. Therefore, there can be no assurance as to
the amount of grants, milestone payments or upfront payments (if
any) that we will receive for any taxable year. Moreover, we may
earn income from sublicensing, which may be passive unless certain
conditions are satisfied. There is no assurance that the Internal
Revenue Service (IRS) will not challenge the classification of any
of our income items for PFIC purposes for any taxable year.
Accordingly, there is no assurance that we will not be a PFIC for
any taxable year under the income test.
In addition, we currently hold, and
expect to continue to hold, a substantial amount of passive assets,
including cash. The average value of our assets (including
goodwill) for purposes of determining our PFIC status for any
taxable year may be determined, in large part, by reference to our
market capitalization, which has fluctuated substantially over time
and may continue to be volatile. Due to the volatility of our
market capitalization, we may be a PFIC under the asset test for
any taxable year if our cash and other passive assets constitute
50% or more of the value of our total assets (including
goodwill).
As discussed in our Annual Reports
on Form 20-F for 2019 and 2020, we were likely a PFIC for our
taxable years of 2019 and 2020. If we were a PFIC for 2019 or 2020,
we will generally continue to be treated as a PFIC with respect to
a US investor who owned our common shares during any portion of
such years, even if we are not a PFIC for 2021 or any other taxable
year, unless the US investor makes a “deemed sale” election with
respect to our common shares.
Although we have not obtained
independent valuations of our assets for our taxable year of 2021
and thus are not in a position to make a definitive determination
as to whether we were a PFIC in 2021, based on the composition of
our income and assets during 2021 and the estimated value of our
assets (which is based on our average market capitalization during
2021), we believe that we were not a PFIC for 2021. However, for
the reasons discussed above there can be no assurance that the IRS
will agree. Because our PFIC status for 2022 or any future taxable
year will depend on the composition of our income and assets and
the value of our assets, we cannot express any expectation
regarding our PFIC status for the current or any future taxable
year.
US investors that hold our common
shares during any taxable year in which we are a PFIC generally
will be subject to adverse US federal income tax consequences,
including (i) the treatment of all or a portion of any gain on
disposition as ordinary income, (ii) the application of a deferred
interest charge on such gain and the receipt of certain dividends
and (iii) the requirement to file certain reports to the IRS. We do
not intend to provide the information that would enable investors
to take a qualified electing fund election that could mitigate the
adverse US federal income tax consequences if we are a PFIC for any
taxable year.
For further discussion, including a
description of purging elections, see “Item 10. Additional
information—Section E. Taxation.”
ITEM 4.
|
INFORMATION ON
THE COMPANY
|
A.
|
History and development of the
company
|
We are a Swiss stock corporation
(société anonyme)
organized under the laws of Switzerland. We were formed as a Swiss
limited liability company (société à responsabilité limitée) on
February 13, 2003 with our registered office and domicile in Basel,
Switzerland. We converted to a Swiss stock corporation
(société anonyme) under
the laws of Switzerland on August 25, 2003. Our Swiss enterprise
identification number is CHE-109.878.825. Our domicile and
registered office is in Ecublens, at the École Polytechnique
Fédérale Lausanne (EPFL) Innovation Park Building B, 1015 Lausanne,
Vaud, Switzerland. Our common shares were admitted to trading on
Nasdaq Global Market on September 23, 2016, and trade under the
symbol ACIU.
Our registered and principal
executive offices are located in Ecublens, at EPFL Innovation Park,
Building B, 1015 Lausanne, Switzerland, our general telephone
number is (41) 21 345 91 21 and our internet address is
www.acimmune.com. Our agent for service of process in the United
States is Cogency Global Inc. located at 122 East 42nd Street, 18th
Floor, New York, New York 10168. Our website, and the information
contained on or accessible through our website, are not part of
this document. The SEC maintains an internet site that contains
reports, proxy and information statements and other information
regarding issuers that file electronically with the SEC, which can
be found at http://www.sec.gov.
Our principal expenditures since
January 1, 2019 have been our research and development expenses, as
more fully described elsewhere in this Annual Report.
AC Immune is a leading, clinical
stage biopharmaceutical company advancing one of the broadest
portfolios focused on pioneering precision medicine for
neurodegenerative diseases. Our highly differentiated approach
integrates novel therapeutics and diagnostics to overcome the
fundamental challenge in this therapeutic area – the high number of
co-pathologies driving disease and the urgent need for more
tailored therapeutic regimens.
Leveraging our dual proprietary
technology platforms, we have built a comprehensive pipeline of
first-in-class or best-in-class candidates spanning multiple
treatment modalities and targeting both established and emerging
neurodegenerative pathologies. We are currently advancing eleven
therapeutic and three diagnostic programs, with seven currently in
clinical trials, targeting five different types of misfolded
pathological proteins related to AD, PD and other neurodegenerative
disorders. Our pipeline assets are further validated by the
multiple partnerships we have established with leading global
pharmaceutical companies. We believe our validated technology
platforms and personalized medicine approach position AC Immune to
revolutionize the treatment of neurodegenerative disease in the way
precision diagnostics and targeted therapies are revolutionizing
the treatment of cancer.
Figure 1: AC Immune
investment highlights
(1) As of December 31, 2021
Our Team
We have assembled an outstanding
management team with relevant scientific, clinical and regulatory
expertise. Our scientific founders, Dr. Jean-Marie Lehn, Dr. Claude
Nicolau, and Dr. Fred van Leuven, are regarded as pioneers in their
respective scientific domains, including in the study of AD. Our
co-founder and Chief Executive Officer, Dr. Andrea Pfeifer, a
Pharmacologist with a Ph.D. in cancer research and a former
National Institute of Health researcher, has a 30-year track record
in product innovation and implementation, and was formerly Head of
Nestlé Global Research and the co-founder of Nestlé Venture Fund.
Dr. Marie Kosco-Vilbois, our Chief Scientific Officer, brings more
than 20 years of experience in various aspects of discovery
research and drug development, including work on multiple drug
development programs. Prof. Johannes Rolf Streffer joined AC Immune
in 2021 as our Chief Medical Officer. Prof. Streffer is a
Neurologist and Psychiatrist with extensive expertise in AD
including biomolecular modalities such as PET, volumetric and
functional MRI, genetics, cognition and cerebrospinal fluid (CSF)
marker.
Unmet need in neurodegenerative
diseases
Figure 2: Neurodegenerative diseases represent a large and growing
market
(1) Alzheimer’s
Disease International; (2) Parkinson’s disease; (3)
Michael J. Fox
Foundation; (4) Limbic-predominant age-related TDP-43
encephalopathy; (5) Nelson PT et
al., Brain 2019; (6) National Institute of Neurological
Disorders and Stroke
Neurodegenerative diseases,
including dementias and other diseases associated with protein
misfolding, are prevalent, but there is currently an absence of
reliable, early-stage diagnosis and disease-modifying treatments
for these diseases. The growth in the number of people with
neurodegenerative diseases has been significant, as evidenced by
the prevalence of people affected by AD and PD, two of the most
common neurodegenerative diseases.
|
• |
The World Health Organization recognizes dementia as a global
public health priority.
Worldwide, there is a new case of dementia every 3 seconds, with an
estimated global patient population of greater than 50 million in
2020. This is predicted to increase to 139 million by 2050
(Alzheimer’s Disease International).
|
|
• |
The estimated total healthcare costs for the treatment of
Alzheimer disease in the United States in 2021 is estimated to be
USD 355 billion per the Alzheimer’s Association, with the worldwide
cost for dementia expected to increase to approximately USD 2.8
trillion annually by 2050 as the population ages. In fact, if the
estimated global costs of dementia were a country, it would be the
14th
largest economy in the world.
|
Current
diagnostic and treatment paradigms for neurodegenerative diseases
are suboptimal. Diagnosis typically takes the form of observation
of cognitive, functional and behavioral impairment and other
symptoms of the diseases, which are generally only apparent after
irreversible neuronal damage has already occurred. Until 2021,
there were five approved therapies for AD, all of which provided
only modest efficacy in treating the symptoms of the disease while
having significant side effect risks and failing to address the
progression of the disease. Despite these shortcomings, marketed
therapies, such as Eisai and Pfizer’s Aricept, have achieved peak
annual global sales of approximately USD 4 billion prior to loss of
exclusivity. Similarly, in the treatment of PD, the current
standard of care is intended only to alleviate physical
symptoms.
On June 7,
2021, the U.S. Food and Drug Administration (FDA) granted
accelerated approval of Biogen’s anti-Abeta monoclonal antibody,
aducanumab, for the treatment of Alzheimer’s disease (AD), making
it the first FDA-approved, potentially disease-modifying therapy
addressing this high unmet medical need. The FDA based its decision
to approve aducanumab on the reduction of Abeta plaques in the
brain as a surrogate endpoint. Biogen will need to complete a large
clinical trial to confirm that removing Abeta plaque has clinical
benefits on cognition and function.
On January
11, 2022, CMS released a proposed National Coverage Determination
decision memorandum: stating it would cover FDA-approved monoclonal
antibodies (including ADUHELM) that target amyloid for the
treatment of Alzheimer’s disease solely for people enrolled in
qualifying clinical trials. A final decision is expected in April
2022.
Neurodegenerative disease
overview
Folding and unfolding of proteins
are important ways of regulating the biological activity and
cellular location of those proteins. Misfolding of proteins occurs
due to a breakdown of cellular quality control systems and is a
common feature of many neurodegenerative diseases. Misfolded
proteins are unable to carry out their normal functions and
aggregate to form insoluble deposits in the brain, which eventually
lead to neuronal damage and cell death. The progression of
neurodegenerative diseases, such as AD and PD, is linked to the
spread of misfolded, pathological protein aggregates throughout the
brain. Figure 3 shows how misfolded proteins play a key role in the
pathology of neurodegenerative diseases.
Figure 3:
Misfolded proteins key impact on the pathology of neurodegenerative
diseases
Typically, protein misfolding
occurs in response to cellular stress, which can be triggered by
many different, largely unknown, causes. A cascade of molecular
events begins with the misfolding of single proteins within a cell,
which then aggregate and ultimately form larger aggregates
including plaques and tangles. These misfolded proteins are then
exported or shed from dying neurons where they can spread to
healthy cells nearby. Once inside, misfolded proteins can interact
with normal proteins and cause them to misfold in a process known
as “seeding,” leading to spreading of the disease pathology
throughout the brain, increased neuronal death and a progressive
decline in cognitive function.
Figure 3 also shows how our
therapies are designed to intervene and prevent key pathological
steps in the progression of neurodegenerative diseases. They are
designed to (i) prevent initial misfolding; (ii) promote
disaggregation of misfolded proteins; (iii) inhibit spreading of
pathological protein to healthy cells; (iv) prevent seeding of new
misfolded protein aggregates inside healthy cells; and (v) inhibit
downstream neurodegeneration. This robust approach to targeting
neurodegenerative diseases is enabled by our two validated
technology platforms, SupraAntigen and Morphomer, which generate
highly specific biologics and small molecule inhibitors that can
distinguish normal from misfolded proteins and inhibit key disease
pathways both inside and outside of cells.
Our strategic vision
Our goal is to continue leveraging
our proprietary discovery platforms, SupraAntigen and Morphomer, to
become a global leader in precision medicine for the diagnosis and
treatment of neurodegenerative diseases. We are executing a clear
business strategy built on three pillars: (i) accelerate
development of novel therapeutics in AD with our partners; (ii)
expand our strategic focus in Parkinson’ disease (PD) and non-AD
neurodegenerative diseases, including NeuroOrphan indications and
limbic-predominant age-related TDP-43 encephalopathy (LATE); and
(iii) a continued focus on diagnostics enabling precision medicine
to be an ultimate differentiator for the Company.
Figure 4: AC
Immune’s three-pillar strategy
(1) Parkinson’s disease; (2) Down syndrome; (3) Neurodegenerative
diseases; (4) Multiple system atrophy; (5) TAR DNA-binding protein
43
Our three-pillar execution
strategy reflects our unique precision medicine approach, which
ultimately creates differentiation due to our ability to address
the high levels of co-pathologies present in AD and other
neurodegenerative diseases. Much like cancer, neurodegenerative
diseases are heterogeneous and may require multiple therapeutic
interventions tailored to patients’ specific disease drivers, to be
used in concert in order to slow or stop the disease course.
Ultimately, it is our belief that precision medicine will increase
the chance of treatment success by enabling clinical trial
participants to be better defined by their various proteinopathies,
affording treatment with the right therapies at the right
time.
AC Immune has
established itself as a leader in developing precision medicines
for neurodegenerative diseases by utilizing our diagnostic
capabilities to enable improved diagnosis of co-pathologies,
patient selection and assessment of clinical trial outcomes. Our
dual technology platforms allow for a multi-modal approach
encompassing a portfolio of vaccines, antibodies and small
molecules tailored to the underlying pathology driving patients’
disease. In addition to generating targeted monotherapies, this
approach creates the potential for combination regimens, which may
treat a broader spectrum of disease and offer greater
efficacy.
AC Immune’s Roadmap to Successful Therapies for
Neurodegenerative Diseases
Precision medicine is a key element
of our six-point framework for developing successful therapies in
neurodegenerative diseases, building on one of the broadest
pipelines in the field.
Figure 5: AC
Immune’s roadmap to successful therapies for neurodegenerative
diseases
(1) Reardon S, Nature 2018; (2)
Pontecorvo MJ, et al.,
Brain 2019; (3) Gordon BA, et al., Brain 2019;
(4) Positron emission tomography; (5) Strydom A, et al., Alzheimer’s Dement
(NY) 2018; (6) Lott IT and Head E.,Nat Rev Neurol.
2019; (7) Down syndrome-related Alzheimer’s disease; (8) TAR
DNA-binding protein 43; (9) Robinson JL, et al., Brain
2018; (10) Heneka
MT et al., Nat Rev Neurosci. 2018; (11) Wang S et al., Int Immunopharmacol.
2019; (12) Monoclonal antibody; (13) Small molecule; (14)
NOD-like receptor protein 3; (15) Apoptosis-associated speck-like
protein containing a CARD, also PYCARD
Treat earlier
Identifying patients at risk or in
early stages of disease when pathological burden is low and
neuronal health is preserved offers the best chance of intercepting
pathological spread in neurodegenerative diseases. For example, it
is now believed that treatments targeting beta-amyloid (Abeta) may
be most effective before symptoms become apparent. The Alzheimer’s
Prevention Initiative (API) trial of crenezumab aims to answer this
fundamental question.
Target Tau
Tau plays a very important role in
neurodegeneration. Understanding whether the aggregation and
spreading of pathological Tau throughout the brain can be stopped
by therapies targeting Tau is a critical question that we are
examining. This is being addressed through AC Immune’s multiple Tau
research programs in early and late-stage diseases.
More homogeneous populations
Multiple pathologies are thought to
contribute to the development of AD, including genetic, lifestyle
and environmental factors. To understand if a candidate drug has
therapeutic potential, it is important to first engage more
genetically homogeneous patient populations to minimize variability
with respect to pathophysiology. We are developing these efforts
with our prevention studies in genetically defined populations such
as familial AD and DS-related AD.
Precision medicine
Building on the understanding that
multiple pathologies contribute to AD, there is a need to
accurately diagnose and target the underlying pathology. We are
developing an integrated diagnostic and therapeutic strategy to
deliver, for the first time, precision medicine for patients with
neurodegenerative conditions.
Target neuroinflammation
It is well established that
microglia maintain a healthy brain environment by clearing debris,
including misfolded and aggregated Abeta, Tau and alpha-synuclein
(a-syn). Chronic hyper-stimulation of microglial cells by these
protein aggregates is now emerging as a hallmark of AD – and
potentially all neurodegenerative diseases – that leads to unwanted
inflammation and further damage to brain cells. We focus on the
NOD-like receptor pyrin domain-containing protein 3 (NLRP3)
inflammasome pathway, based on emerging evidence showing its
particular relevance for neurodegenerative diseases.
Non-AD indications
We are also broadening our
strategic activity in other neurodegenerative diseases such as
Parkinson’s disease and frontotemporal dementia with the genetic
microtubule-associated protein tau (MAPT) mutation – a NeuroOrphan
disease we aim to address with our Morphomer Tau small molecule
aggregation inhibitors. Finally, we will also continue to advance
our suite of potentially best-in-class diagnostics, particularly
those for Parkinson’s disease and TDP-43-based pathologies.
Key elements of our approach
include:
1. Execution
on advancing our product candidates, in partnership or alone, from
clinical development to regulatory approval and potential
commercialization
Figure 6: Our broad and robust clinical stage
pipeline
(1) Alzheimer’s disease; (2) Open label extension study is ongoing;
(3) Positron emission tomography; (4) Progressive supranuclear
palsy; (5) Prevention trial API-ADAD in Colombia; (6) Down
syndrome-related Alzheimer’s disease; (7) alphasynuclein; (8)
Parkinson’s disease; (9) Multiple system atrophy
Our clinical stage product
candidates include:
|
• |
ACI-35.030.
Janssen and AC Immune are evaluating the anti-phosphorylated-Tau
(anti-pTau) vaccine candidate ACI-35.030 in a Phase 1b/2a study in
subjects with early AD. Interim results show that ACI-35.030
vaccination generated a strong antigen-specific antibody response
against pTau in 100% of participants, achieving anti-pTau antibody
levels of about two orders of magnitude higher than pre-vaccination
levels, whereas anti-ePHF (enriched paired helical filaments)
antibody titers increased by one order of magnitude from baseline
as early as two weeks after the second injection at week 8 of the
mid-dose of ACI-35.030. No clinically relevant safety concerns
related to the vaccine candidate were observed. Based on these
results, the second highest dose cohort was expanded in Q2 2021 to
facilitate plans for further late-stage development. ACI-35.030
specifically targets pathological pTau species and is eventually
intended as a disease-modifying treatment for AD and other
Tauopathies.
|
|
• |
ACI-24
for AD. A first Phase 1/2 study was completed and finalized
in 2019. The subsequent Phase 2 study in AD assessed the safety,
tolerability, immunogenicity and target engagement of ACI-24 using
intramuscular immunizations and analyzed the effects of ACI-24 on
brain amyloid as assessed by PET imaging. This trial was completed
and finalized in November 2021. ACI-24 was safe and well tolerated
and triggered a clear IgM response with lower Abeta-specific IgG
titers. While no apparent effect in amyloid-PET was observed in
this limited study population, there was evidence of a
pharmacodynamic effect observed by an increase of CSF Aβ1-40 and
Aβ1-42 levels compared to the placebo, thus suggesting target
engagement. These results support the clinical development of the
optimized formulation of ACI-24 (i.e. ACI-24.060) with Abeta
unrelated T-helper cell epitopes to increase the magnitude and the
boost-ability of the antibody response.
|
|
• |
ACI-24
for DS. Our Phase 1b clinical study of ACI-24 for
individuals with DS, intended to assess safety, tolerability and
immunogenicity at two doses, was completed and results reported in
Q1 2021. The results support a favorable safety and tolerability
profile of ACI-24 and show a pharmacodynamic response in this
vulnerable patient population and the advancement of this program
with the optimized formulation of ACI-24. The Clinical Trial
Application (CTA) for the next study evaluating the optimized
formulation of ACI-24 in AD and Down syndrome populations was
submitted in Q4 2021. The trial initiation is planned in H1
2022.
|
|
• |
ACI-7104. ACI-7104,
the optimized formulation of the clinically-validated PD vaccine
candidate PD01, will advance into an adaptive, biomarker-based
Phase 2 study. This trial will evaluate an initial dose-response of
the optimized formulation focusing on immunogenicity against a-syn
and pathological a-syn species. Additionally, the identification or
verification of disease-specific biomarkers and progression of
motor and non-motor symptoms of Parkinson’s disease will be
monitored, together with digital, imaging and fluid biomarkers, in
the second part of the study. The trial initiation is planned in H2
2022.
|
|
• |
Semorinemab. Our
collaboration partner, Genentech, a member of the Roche Group,
completed a first Phase 2 study (Tauriel) conducted in patients
with prodromal-to-mild AD in Q3 2020. This trial did not meet its primary efficacy endpoint
of reducing decline on Clinical Dementia Rating-Sum of Boxes
(CDR-SB) compared to placebo; the primary safety endpoint was met.
A second Phase 2 study (Lauriet) conducted in patients with
mild-to-moderate AD was completed in Q3 2021 and top-line data from
showed a statistically significant reduction on one of two
co-primary endpoints, ADAS-Cog11. The second co-primary endpoint,
ADCS-ADL, and secondary endpoints were not met. Safety data showed
that semorinemab is well tolerated with no unanticipated safety
signals. Genentech reported that the open label portion of the
study will continue as planned and that further analyses are
ongoing. Semorinemab is designed to slow the prion-like propagation
of Tau pathology, which coincides with both clinical symptoms and
disease progression in AD.
|
|
• |
Crenezumab. Roche
announced in 2019 the discontinuation of the Phase 3 clinical
trials in AD but is continuing in a landmark prevention trial in
Colombia, in a population of genetically predisposed people at risk
of developing familial AD. The overall beneficial safety profile
was confirmed in the CREAD studies, supporting use of crenezumab in
healthy individuals with risk of developing AD. Top-line results
from this Phase 2 Prevention trial are expected in H1 2022.
|
|
• |
Morphomer
Tau aggregation inhibitors. In collaboration with our
partner, Lilly, we are researching and developing small molecule
Tau aggregation inhibitors with plans to evaluate candidates in AD
and NeuroOrphan indications. We completed a Phase 1 clinical study
in healthy volunteers with ACI-3024, in Q2 2020, which showed a
dose-dependent exposure and brain penetration, achieving the
desired levels of ACI-3024 in the CSF. In addition to AD, the
program was expanded to NeuroOrphan indications and ACI-3024 will
be further evaluated for efficacy in models of rare Tauopathies.
Continued candidate characterization across the research program
has also identified new and highly differentiated candidates with
excellent cerebrospinal fluid exposure and selectivity for
pathological aggregated Tau. These will be broadly developed in
Tau-dependent neurodegenerative diseases.
|
|
• |
Tau-PET
tracer. PI-2620 is our Tau-PET imaging agent. We are working
with our partner, LMI, to advance PI-2620 as a highly
differentiated, best-in-class Tau diagnostic for AD as well as
non-AD Tauopathies such as progressive supranuclear palsy (PSP) and
corticobasal degeneration (CBD). Results have demonstrated
PI-2620’s differentiated characteristics as a diagnostic tool for
studying Tau-related diseases. PI-2620 completed a Phase 2 clinical
trial in AD in Q4 2021.
|
A study published in Movement
Disorders indicated a value of PI-2620 for evaluating corticobasal
syndrome, providing quantitatively and regionally distinct signals
in beta-amyloid-positive as well as beta-amyloid-negative
corticobasal syndrome. Further, results demonstrated PI-2620’s
excellent characteristics as a diagnostic tool for studying
Tau-related diseases following a recent publication (J Cereb Blood
Flow Metab) that PI-2620 binding characteristics in cortical
regions differentiated between 3/4R- and 4R-tauopathies and might
serve as a supportive readout in the diagnostic workup of
neurodegenerative disorders. Two test-retest studies in PSP (Phase
1) are open and recruiting with results anticipated in H2
2022.
|
• |
A-syn-PET
tracer. Our next-generation PET imaging tracer, derived from
our Morphomer platform, has shown significant potential to reliably
detect and map deposits of pathological alpha-synuclein protein in
the brain. Supported by the Michael J. Fox Foundation for
Parkinson’s Research (MJFF), a first-in-human study and an
investigator-initiated study of our latest diagnostic agent
targeting a-syn were initiated in Q1 and Q3 2021, respectively. The
readouts of these trials in patients with PD, multiple system
atrophy (MSA) and other synucleinopathies are anticipated by Q2
2022.
|
2. Expand
product development into NeuroOrphan and additional
neurodegenerative diseases
Beyond AD, we
aim to pursue additional neurodegenerative diseases such as
Parkinson’s disease (PD) and NeuroOrphan indications, specifically
Tau-, a-syn- and TDP-43-driven diseases, such as FTLD-Tau (e.g.,
PSP, CBD, FTLD-MAPT), MSA, and ALS and FTLD-TDP, respectively. As
part of this planned strategic move, AC Immune acquired certain
a-syn assets from Affiris in 2021, gaining an advanced, clinical
stage and validated a-syn vaccine candidate for development against
PD in the process.
Pursuing
NeuroOrphan indications may enable us to obtain a streamlined
regulatory approval pathway and favorable reimbursement for any
approved products. In addition, we are accelerating our novel
therapeutic and diagnostic candidates targeting a-syn as a primary
pathology in Parkinson’s disease and other a-synucleinopathies. See
below for a summary of our early-stage diversified novel targets
pipeline including non-AD neurodegenerative diseases, with an
in-house focus on NeuroOrphan indications.
Figure 7:
Robust novel targets pipeline: diversification into non-AD and
non-CNS diseases
(1) Parkinson’s disease; (2) TAR DNA-binding protein 43;
(3) Limbic-predominant age-related TDP-43 encephalopathy; (4)
Positron emission tomography;
(5) NOD‑like receptor protein 3; (6)
Apoptosis-associated speck-like protein containing a CARD, also
PYCARD
|
3. |
Accelerating
the advancement of our diagnostic portfolio
|
Early detection of
neurodegenerative diseases may be critical to enhancing the
effectiveness of both symptomatic and disease-modifying therapies.
As a result, therapeutic development for AD increasingly focuses on
treating early-stage disease to delay or prevent progression and to
preserve the maximum amount of cognitive function before it is
irreversibly lost. Most clinical studies now target mild or even
preclinical stages of the disease increasing the need for accurate
diagnosis that is independent of potentially subjective cognitive
metrics. At least one study estimates that as many as one-third of
patients in previous AD studies did not in fact have AD. Accurate
and early diagnosis of AD is thus a substantial unmet market need,
and diagnostic products will have a key role in generating a new
treatment paradigm, including by selecting more uniform and
stage-specific clinical study subjects, tracking patient progress
and results, managing patients who are receiving treatment, and
ultimately diagnosing disease at its earliest stage for immediate
treatment.
Figure 8: The
need for precision medicine in AD: improved clinical trials,
diagnosis and treatment of neurodegenerative diseases
(1) Alzheimer’s disease; (2) Neurodegenerative diseases; (3)
TAR DNA-binding protein 43; (4) Alpha-synuclein; (5) Intermediate
level of Alzheimer’s disease neuropathological change; (6) High
level of Alzheimer’s disease neuropathological change
Ref: Adapted from Robinson
et al., Brain, 2018
We are developing a suite of
companion diagnostics designed to be first-in-class or
best-in-class, which will enable improved diagnosis of
co-pathologies, patient selection and assessment of clinical trial
outcomes. We currently have four diagnostic programs in our
pipeline, developed using our proprietary technology platforms and
targeting the therapeutic targets: Tau, a-syn and TDP-43.
Leveraging our Morphomer platform,
we are also developing proprietary PET imaging diagnostics for
diseases resulting from the misfolding of a-syn and TDP-43
proteins. No such diagnostics are currently available for these
important pathologies and AC Immune has identified promising
compounds with high affinity and target specificity, as well as
favorable central nervous system (CNS) pharmacokinetic properties.
In 2020, the a-syn-PET tracer won the Ken Griffin Alpha-synuclein
Imaging Competition from The Michael J. Fox Foundation for
Parkinson’s Research. Our novel TDP-43-PET tracer and our
antibody-based immuno-assay for biofluid detection of TDP-43 also
were awarded highly competitive grants from the EU Joint Programme
– Neurodegenerative Disease Research’ (JPND) and The Target ALS
Foundation, respectively, in 2020. Our diagnostics for a-syn and
TDP-43, if validated clinically, could become the first in the
world to effectively diagnose these proteinopathies, which are
highly relevant for multiple neurodegenerative diseases.
|
4. |
Continuing to
optimize our long-term growth by selectively partnering product
candidates for global development and commercialization
|
We have a strong track record of
establishing value-driving collaboration agreements with leading
pharmaceutical companies, including two collaborations with
Genentech, one with Janssen and one with Lilly. This strategy
allows us to leverage our partners’ scientific, development,
manufacturing and commercialization expertise and other resources
while partially monetizing our investments, de-risking and
accelerating the development of our product candidates. This
strategy also enables us to use non-dilutive partnership revenue to
bolster our investment into our early-stage proprietary programs
and fuel our continued growth. We have five current collaboration
agreements with leading global pharmaceutical companies, summarized
in the table below:
Figure 9:
External validation and cash generation through external
collaborations1
(1) Disclosure limited due to confidentiality agreements with
collaboration partners; (2) in millions; (3) Positron emission
tomography; (4) Phase 1 completed; (5) Equity investment; (6)
Converted to CHF on date of receipt; (7) Excludes convertible note
agreement of USD 50 million
For any additional product
candidates targeting large markets, we may, if appropriate,
selectively partner with leading companies that we believe can
contribute development, manufacturing and marketing expertise,
geographic reach and/or other resources that can enhance the value
of our wholly-owned products. We will continue to seek to retain
certain indications (e.g., NeuroOrphan) and/or geographies, such
that we can begin to grow our own marketing capabilities as we
develop AC Immune into a fully integrated pharmaceutical
company.
Additionally, in this respect, we
established a strategic partnership with WuXi Biologics for its
expertise in manufacturing biologicals in China and potential
collaborations regarding AC Immune’s SupraAntigen platform.
The benefits of our
clinically-validated, proprietary technology platforms
The engines that drive our growth
are our two unique proprietary and versatile technology platforms:
our SupraAntigen platform, which is our biological and
immunological platform, and our Morphomer platform, which is our
chemical platform. These platforms generate biologics (vaccines and
antibodies) and small molecules, respectively, which are designed
to selectively interact with the misfolded proteins that are common
in a broad range of neurodegenerative diseases. These
clinically-validated platforms form the basis of our ongoing
pipeline development and the value-driving strategic partnerships
we have established to date.
The key aspect of both our
SupraAntigen and Morphomer technology platforms is conformational
specificity, which we believe is central to the development of
effective and safe therapeutics for neurodegenerative diseases. Our
SupraAntigen platform targets misfolded proteins through antigens
displayed on the surface of liposomes, which mimic the targeted
pathological form of the protein. In a complementary approach, our
Morphomer platform uses small molecular weight compounds to target
the aggregation and seeding process, which prevents the misfolded
proteins from aggregating inside the cell and prevents the
formation of new misfolded proteins in healthy neighboring cells
through a seeding mechanism. Small molecules derived from our
Morphomer platform, which we refer to as Morphomers, not only
inhibit aggregation of pathological proteins, but also promote
disaggregation of already formed aggregates, thereby potentially
enhancing their therapeutic potential even in established disease
states.
Figure 10:
Morphomer and SupraAntigen platforms: an integrated approach to
CNS1-specific therapies
(1) Central nervous system; (2) Blood-brain barrier; (3)
Positron emission tomography
The SupraAntigen platform was first
developed by AC Immune’s scientific co-founders to overcome a
challenge common to neurodegenerative diseases: the lack of
immunogenicity of disease-causing self-proteins. The SupraAntigen
platform uses liposomes (small spherical vesicles formed by a lipid
bilayer) to present specific antigens designed to evoke an immune
response. SupraAntigen is used to generate conformation-specific
antibodies for immunotherapy in neurodegenerative diseases. The
overarching idea behind the platform is that antibodies, which are
large in size, are well-suited to target extracellular proteins,
interrupt spreading of pathological proteins, and break up and
clear aggregates of misfolded proteins through phagocytosis.
AC Immune has acquired advanced
mastery of the design and manipulation of liposomes to develop
either passive or active immunization techniques to generate
antibodies targeting neurodegenerative diseases. When pursuing
active immunization approaches, we use liposomes carrying a
specific antigen as a vaccine. After vaccination with a liposome,
antigen and confirmation-specific antibodies are produced naturally
by the host with very high affinity without further optimization.
This immune response can be long-lasting and may be ideal to
prevent the onset of a disease, as the immune system is now primed
to rapidly identify disease-causing misfolded proteins.
Product candidates generated
utilizing the SupraAntigen platform include vaccines ACI-35 in
Phase 1b/2a for AD and ACI-24 in Phase 2 for AD and Phase 1b for
DS, as well as the antibody crenezumab in Phase 2 for AD and the
preclinical candidates targeting a-syn and TDP-43 for PD and
NeuroOrphan indications.
The Morphomer platform is designed
to enable the development of small molecules (Morphomers) able to
bind/interact with beta-sheets containing fibrillary aggregates
from candidate selection through preclinical proof-of-concept.
Morphomers can target pathological protein aggregates in any brain
compartment and are equally well suited for therapeutic and
diagnostic applications.
The first key component of the
Morphomer platform is its library of rationally designed,
CNS-optimized non-dye compounds. AC Immune’s extensive know-how has
enabled the identification of CNS compounds that penetrate the
brain and demonstrate high selectivity for the target. This
knowledge has been used to focus the Morphomer library to
approximately 15,000 compounds that display these favorable
characteristics, making this library an ideal starting point when
developing molecules to target human proteinopathies of the CNS.
Thus, rather than using the non-directed trial and error strategy
of the typical drug development process, the Morphomer platform
utilizes its bias for successful CNS candidates to improve
efficiency and accelerate the early stages of the drug development
process. Extensive expertise in medicinal chemistry and a suite of
proprietary assays developed to screen and validate candidate
compounds enables AC Immune to rapidly optimize multiple, highly
diversified lead compounds for further preclinical and clinical
development.
Therapeutic product candidates
generated by the Morphomer platform include our lead Morphomer Tau
candidates, Morphomer a-syn in PD (preclinical stage) and the
diagnostic programs PI-2620 in Phase 2 and Phase 1 in AD and PSP,
respectively, and a-syn-PET in Phase 1 clinical trial in PD, MSA
and other synucleinopathies and TDP-43-PET imaging agents in the
preclinical stage.
Shifting the current treatment
paradigm for neurodegenerative diseases
Modifying the progression of the
disease requires targeting the specific underlying biological
processes that drive disease progression. Unfortunately, these
processes evolve over the course of many years prior to
manifestation of symptoms and a high percentage of neurons may be
lost prior to clinical manifestation. Earlier intervention or
prevention of the disease could have a major impact, but it
requires accurate disease detection prior to developing symptoms.
Due to recent advancement in biomarker research, people at risk of
developing AD can be diagnosed 10-20 years before symptoms occur,
opening a completely new market segment for the prevention of NDD
when active vaccination will play an important role. This early,
and potentially preventative, precision medicine approach may
ultimately lead to better disease management for patients with
neurodegenerative diseases.
Figure 11:
Treatment and diagnosis of AD
Due to the high level of
co-pathologies involved in neurodegenerative diseases, future
treatment paradigms may involve different combinations of disease
modifiers at various stages of a disease. Therefore, combination
therapies may include combinations of immunotherapies or
combinations of small and large molecules targeting proteinopathies
and neuroinflammation. Our therapeutic product candidates seek to
modify the course of AD by intervening at an earlier stage of the
disease progression, prior to irreversible neuronal damage. Beyond
AD, we believe that we can leverage our proprietary platforms to
generate and employ molecules that address the pathologies of other
neurodegenerative diseases (Figure 12).
Figure 12:
Market opportunities targeting key primary and co-pathologies
(1) Alzheimer’s Association; (2) (NOD)-like receptor protein; (3)
Apoptosis-associated speck-like protein containing a CARD, also
PYCARD; (4) GBD 2016 Parkinson’s Disease Collaborators Lancet
Neurology 2018; (5) Limbic-predominant age-related TDP-43
encephalopathy; (6) Nelson et. al. Brain 2019; (7) TAR DNA-binding
protein 43; (8) National Institute of Neurological Disorders and
Stroke (NINDS) Progressive Supranuclear Palsy Fact Sheet; (9) NINDS
Multiple System Atrophy Fact Sheer; (10) ALS Association Rare
Disease 2013; (11) NINDS Amyotrophic Lateral Sclerosis Fact Sheer;
(12) Knopman and Roberts J. Mol. Neurosci. 2011
In support of shifting the current
treatment paradigm from treatment to prevention, we are the leader
in discovering new PET imaging agents to improve the timing and
accuracy of diagnoses in neurodegenerative diseases. In our
pipeline, we have three families of diagnostic candidates that were
developed through our Morphomer platform, which target Tau, a-syn
and TDP-43. We believe our Tau-PET imaging program has received
external validation through our partnership with LMI, a leader in
imaging agents. We are also developing a-syn and TDP-43 PET imaging
agents for PD and other neurodegenerative diseases.
With our unique integrated approach
focused on precision medicine, we believe that our diagnostic
product candidate pipeline will complement our disease-modifying
treatment product candidate pipeline and potentially reshape the
clinical course and treatment of neurodegenerative diseases.
Our clinical programs
Anti-pTau vaccine
In collaboration with Janssen, we
are advancing an anti-pTau vaccine program directed against a key
component of the pathology of AD: phosphorylated Tau proteins,
found in Tau tangles. Developed using our SupraAntigen technology,
our vaccine is designed to stimulate a patient’s immune system to
produce antibodies against misfolded and phosphorylated,
pathological Tau protein, which aggregate to create the
neurofibrillary tangles that characterize AD.
Advantages of Tau vaccination over
other therapeutic approaches
Tau vaccines which are able to
induce a long-lasting and boost-able antibody response have the
potential to be even more advantageous than other anti-Tau
therapeutic modalities such as small molecules or monoclonal
antibodies, which typically show much shorter half-lives
in vivo, requiring more
frequent administration. Tau vaccines such as ACI-35.030 may thus
offer a more cost effective, and less invasive approach for the
treatment or prevention of Tau pathology, which may be particularly
relevant for addressing slow-progressing chronic neurodegenerative
Tauopathies such as AD.
ACI-35
ACI-35 is an initial formulation
liposomal anti-pTau active investigational vaccine designed to
elicit antibodies against extracellular pTau protein in order to
prevent and reduce the spread and development of Tau pathology
within the brain. In preclinical testing, the vaccine candidate
induced an antibody response that was highly specific to
phosphorylated Tau. This antibody response resulted in a
significant reduction of pTau and in animal disease model. ACI-35
was the first vaccine candidate against pathological pTau to be
tested in a clinical study involving patients with mild-to-moderate
AD. The Phase 1b study was completed in June 2017.
Mechanism of
action
|
• |
ACI-35 is composed of a human pTau synthetic peptide T3 as the
antigen, derived from Tau sequence 393-408 and phosphorylated at
serine residues S396 and S404. Lipidation of the peptide enables it
to embed itself into the lipid bi-layer of the liposome and confers
a specific conformation to the peptide (Theunis et al., PLoS ONE 2013).
|
|
• |
In wild-type and transgenic mice, immunization with ACI-35
generated a specific antibody response to phosphorylated vs.
non-phosphorylated Tau protein (Vukicevic et. al. AAT-AD/PD 2020).
|
|
• |
In transgenic mice, immunization with ACI-35 led to a
significant decrease of soluble and insoluble total Tau protein and
insoluble pTau species in brain (Figure 13).
|
Figure 13:
Immunization of hTauP301L mice leads to a reduction of the levels
of pS396 and HT7 in the Sarkosyl insoluble Tau fraction of the
forebrain of Tau.P301L mice
Ref: Theunis et al., PLoS ONE 2013
Clinical
development
Phase 1b study
design
The safety, tolerability and
immunogenicity of ACI-35 were tested in a Phase 1b study in
participants with mild-to-moderate AD. It was a randomized,
placebo-controlled, double-blind study. Different dosages and
dosing schedules were investigated in an ascending dose design.
Multiple injections of ACI-35 were administered per cohort for
active or placebo treatment in a three-to-one ratio.
Safety
The ACI-35 vaccine was considered
to be safe and well tolerated with no events related to CNS
inflammation. Five SAEs were observed in three participants.
Antibody
response
ACI-35 elicited a rapid induction
of anti-pTau antibodies after the first immunization in all study
cohorts, indicating a T-cell-independent antibody response.
However, this response lacked the boosting effect desired for
optimal long-term and potentially preventive application.
Therefore, in a collaborative effort, AC Immune and Janssen are
developing two optimized anti-pTau vaccine candidates, ACI-35.030
and JACI.35.054, which are currently being tested in early AD
subjects in a Phase 1b/2a study.
ACI-35.030
ACI-35.030 is an optimized
liposomal anti-pTau vaccine formulation designed to elicit an
enhanced antibody response. In preclinical studies, ACI-35.030
showed that it retains the excellent non-clinical safety profile
and the highly specific antibody response against pTau observed
with ACI-35, while demonstrating an enhanced and more uniform
antibody response compared to first generation ACI-35. We are
developing ACI-35.030 with Janssen in accordance with our
collaboration agreement.
Mechanism of
Action
|
• |
ACI-35.030 comprises a pTau peptide and a T-cell epitope
capable of binding to human leukocyte antigen-major
histocompatibility complex, class II (HLA-DR) molecules.
|
|
• |
In rhesus monkeys, ACI-35.030 induced a specific response to
pTau over non-phosphorylated Tau, similar to that observed with
ACI-35 (Vukicevic et. al.
AAT-AD/PD 2020). This is meaningful as Tau hyper-phosphorylation is
considered an early event in the development of Tau pathology,
occurring even several decades before the onset of Tau
deposits.
|
|
• |
Sera from rhesus monkeys immunized with ACI-35.030 binds
specifically to pathological Tau in brain sections with AD as
compared to healthy human brain tissue (Kosco-Vilbois, KOL event
‘Untangling’ Tau Pathology to Treat Alzheimer’s and
Neurodegenerative Diseases NYC, Nov 2019)
|
|
• |
In preclinical studies, immunization of non-human primates
(NHPs) with ACI-35.030 lead to enhanced and more uniform
anti-pTau IgG -specific antibody titers with boosting effect
compared to ACI-35 (Figure 14).
|
Figure 14:
pTau-specific IgG titers in NHP induced by ACI-35.030 and
ACI-35
Ref: AC Immune, CTAD 2021.
JACI-35.054
JACI-35.054 is an alternative pTau
vaccine, which is developed with Janssen in accordance with our
collaboration agreement. In preclinical studies, JACI-35.054 showed
good safety, and induced a strong antibody response against
pTau.
Mechanism of
action
|
• |
JACI-35.054 is an alternative anti-pTau vaccine comprising a
pTau peptide antigen conjugated to an immunogenic carrier protein
CRM197, combined with adjuvants
|
|
• |
CRM197 is a well-defined recombinant protein that is a
commercially available version of a non-toxic mutant of diphtheria
toxin (DT) A chain and has been shown to be a safe carrier protein
in commercial prophylactic vaccines and clinical trials for a
plethora of different vaccine candidates.
|
|
• |
Immunization of rhesus macaques with JACI-35.054 generates an
antibody response that binds to pathological Tau structures in
human AD brain.
|
Clinical
development
Phase 1b/2a
study
The Phase 1b/2a study is a
randomized, multicenter, double-blind, placebo-controlled clinical
study with a primary objective to assess the safety, tolerability
and immunogenicity of different dosages of ACI-35.030 and
JACI-35.054 in participants with early AD. Secondary objectives
assess additional immunogenicity parameters, while exploratory
endpoints include notable biomarkers of progression of AD as well
as clinical assessments. This Phase 1b/2a study evaluating
ACI-35.030 and JACI-35.054 was initiated in Q3 2019 and is
currently ongoing.
Safety
As of February
11, 2022, 55 subjects have been randomized in the Phase 1b/2a
study, of which 39 subjects are randomized into the Cohort 1 (low-,
mid-, or high- dose levels of ACI-35.030 or placebo), and 16
subjects are randomized into the Cohort 2 (low- or mid-dose levels
of JACI-35.054 or placebo). The active/placebo ratio is 3:1 in each
Cohort. In this study, six SAEs have been reported to date. Each of
these events are considered unlikely related to the study
treatment. These events are: one episode of acute diverticulitis
and one case of sick sinus syndrome in the low-dose level with
ACI-35.030 or placebo; one case of flare of diverticular disease in
one subject, and one left thrombosed popliteal aneurysm and one
right popliteal aneurysm in another subject in the high-dose level
with ACI-35.030 or placebo; and one case of lumbar disc prolapse
leading to hospitalization for surgery in the low-dose level with
JACI-35.054 or placebo.
Antibody response (interim)
Based on interim results from
the first two dose-level sub-cohorts, in all patients after the
first injection, ACI-35.030 treatment led to the strong induction
of antibodies specific for pathological forms of Tau such as pTau
and its aggregated form, enriched paired helical filaments (ePHF).
Anti-pTau IgG titers increased by two orders of magnitude from
baseline two weeks after the first injection of the mid-dose of
ACI-35.030. Anti-ePHF IgG titers increased by approximately one
order of magnitude from baseline as early as two weeks after the
second injection at week 8 of the mid-dose of ACI-35.030. The
geometric mean of the anti-ePHF IgG response was boosted following
additional doses at weeks 8 and 24. The ACI-35.030-induced immune
response was lasting over an initial period of 26-weeks and showed
class-switching from IgM to IgG. Interim safety data further
support ACI-35.030’s favorable safety and tolerability profile,
with no clinically relevant safety concerns related to the study
vaccine observed to date. These results were presented at the CTAD
conference in 2021.
Figure 15:
ACI-35.030 generates a potent Ab1
response6
against pathological Tau
ACI-35.030
generates strong Ab responses against pTau2
in an older population
(1) Antibody; (2) phosphorylated Tau; (3) Enriched paired
helical filaments; (4) at Weeks 2 and 10; (5) at Week 10; (6)
Responders were defined as subjects with an antibody response
higher than a positivity threshold, i.e., a pretreatment value
(baseline antibody titer), multiplied by a threshold factor
(>-2x)
Ref: AC Immune, CTAD 2021.
ACI-24
The original formulation of ACI-24
is an anti-amyloid-beta vaccine candidate that was assessed in AD
and in Down syndrome and was shown to be safe and well tolerated
along with preliminary evidence of immunogenicity and
pharmacodynamic effects in these 2 study populations.
|
• |
ACI-24 consists of an antigenic peptide (Pal1-15) containing
the amino acid sequence 1-15 of the human Abeta1-42 protein, and an
adjuvant, formulated as liposomal suspension. Pal1-15 is presented
on the surface of the liposomes in a conformational format
mimicking the pathological form of the protein which is recognized
by the immune system in order to induce antigen-specific antibody
responses against the pathological forms of Abeta. Preclinical data
demonstrated significant activity in plaque reduction and memory
restoration. ACI-24 formulations have a favorable safety profile,
characterized by a lack of observed local and CNS inflammation and
a mechanism of action independent of inflammatory T cells. ACI-24
formulations are fully owned by AC Immune and have been developed
in-house.
|
Clinical
development in AD
Phase 1/2
Phase 1/2
study
The Phase 1 part of the combined
Phase 1/2 study is completed, and the clinical study report was
finalized in 2019. The efficacy, tolerability and immunogenicity of
ACI-24 were tested in patients with mild-to-moderate AD with four
different doses in a randomized, placebo-controlled, double-blind
study. The different doses were tested via an ascending dose design
in four consecutive cohorts with 12 patients each (nine on active,
three on placebo treatment). ACI-24 was administered with multiple
injections per cohort. The initial safety follow-up period for two
years was shortened to one year mainly for the patients of the last
cohort.
Safety and
tolerability
Due to the observed favorable
safety profile, the treatment-free safety follow-up period of the
Phase 1 part of the study was shortened to one year using a
protocol amendment. Fifteen non-drug related SAEs were observed in
the Phase 1/2 study. This ACI-24 vaccine was considered safe and
well tolerated.
Antibody responses were observed
only in the two higher-dose groups of cohorts 3 and 4, indicating a
dose-dependent effect of the vaccine. No IgG antibody response was
observed in placebo-treated patients of those cohorts.
PET Imaging and
cognitive measures
Although the
study was not powered to examine efficacy, a tendency for reduction
in accumulation in brain amyloid measured by PET imaging was
observed in cohorts 3 and 4. This was paralleled by trends in
functional improvements as measured by CDR-SB and MMSE at week 52
(MMSE 20-28).
Due to the
safety profile and potential dose-dependent reduction of amyloid
plaques as measured by PET imaging, this program was advanced into
a Phase 2 clinical trial. In order to optimize the immune response,
the route of administration was switched to intramuscular, as this
route was associated with a better antibody response in a
non-clinical study.
Phase 2
Phase 2 study design
The Phase 2 double-blind,
randomized, placebo-controlled adaptive design study assessed the
safety, tolerability, immunogenicity and target engagement of
ACI-24 formulations in patients with mild AD. It was conducted in
several European countries and the first dosing occurred in October
2018 via the intramuscular route of administration. The full study
results were presented at CTAD 2021.
Safety and
tolerability
Seven SAEs considered unlikely
related or unrelated to study drug were reported in this study in 5
randomized subjects; 3 of the subjects were in the 1000 μg ACI-24
group (Covid-19 infection; pneumonia; and foot deformity) and 2
subjects were in the placebo group (transient ischemic attack; and
2 episodes of urinary retention and one concussion occurring in one
subject). No episodes of ARIA-E or CNS inflammation were reported
by MRI. ACI-24 was considered safe and well tolerated.
Antibody
response
The vaccine triggered an IgM
response with lower Abeta-specific IgG titers.
Pharmacodynamic
and clinical measures
While no apparent effect in
amyloid-PET was observed in this limited study population, evidence
of a pharmacodynamic effect was shown by an increase of CSF Aβ1-40
and Aβ1-42 levels vs placebo, thereby suggesting target engagement.
No consistent changes in cognitive and other clinical scales were
observed over time, though it should be noted that the study was
not powered for these endpoints.
Overall, these results support
the clinical development of an optimized formulation of ACI-24 with
T-cell help to improve the magnitude and the boostability of the
antibody response. A CTA submission for the next study with the
optimized formulation of ACI-24 in AD and Down syndrome populations
was completed in Q4 2021.
ACI-24 development in Down
syndrome-related AD
The AD dementia that commonly
develops in people with DS bears remarkable clinical and
pathological similarity to familial and sporadic forms of AD and is
characterized by progressive changes in Abeta and a number of other
relevant biomarkers. AC immune is pioneering the development of
anti Abeta vaccine for these individuals and this may also apply to
other populations with genetic predisposition to AD and ultimately
to broader sporadic AD patients.
Individuals with DS have an extra
copy of chromosome 21, which is where the gene for amyloid
precursor protein (APP) resides. These individuals develop AD at a
rate that is three to five times that of the general population and
develop the disease at a much younger age. At autopsy, AD pathology
has been reported in 80% of people with DS over the age of 40 and
100% over the age of 60 years. The prevalence of AD in people with
DS is more than 50% over the age of 50 and 75–100% over the age of
60 years (Strydom, 2018). It is estimated that there are six
million people with DS worldwide, with 250,000 in the US.
Preclinical results published by AC Immune in collaboration with
Dr. Mobley of the University of California, San Diego in March
2016, showed, in a DS mouse model (Ts65Dn), a significant 20%
memory improvement and a 27% reduction of Abeta in the brain
following vaccination with ACI-DS-01, the mouse equivalent of
ACI-24.
Phase 1b
Phase
1b study design
A Phase 1b clinical trial was
completed in 2020 and evaluated the safety and tolerability of
ACI-24, its effect on induction of antibodies against Abeta and
changes in biomarkers such as Abeta levels in blood and CSF, in
adult participants with DS. The study was primarily funded by the
Company with additional partial funding provided by a grant from
the US National Institute on Aging, a part of the US National
Institutes of Health (NIH) and an additional grant from the LuMind
Research Down Syndrome Foundation. This dose-escalation study
included 16 participants across all cohorts, aged 25−45 years and
treated for 12 months, with a 12-month safety follow-up.
Phase
1b results
Study treatment compliance was 100%
and ACI-24 was safe and well tolerated in adults with DS, with no
serious adverse events (SAEs) or evidence for CNS inflammation,
meningoencephalitis, or ARIA. There have been no early subject
withdrawals at any dose during the treatment period. ACI-24
vaccination in adults with DS resulted in encouraging
immunogenicity (generation of anti-Abeta antibodies) and a positive
pharmacodynamic response as measured by an increase in plasma
Abeta. These results support further clinical development of in
DS-related AD with the optimized formulation of ACI-24.
ACI-24.060
AC Immune has developed an
optimized anti-Abeta vaccine formulation, ACI-24.060, which
demonstrated encouraging safety and superior immunogenicity results
in mouse and non-human primate (NHP) studies. Like ACI-24,
ACI-24.060, which contains T-cell help epitopes, has been developed
using our SupraAntigen platform and is designed to stimulate a
patient’s immune system to produce antibodies that specifically
target the misfolded Abeta conformer to prevent plaque accumulation
and to enhance plaque clearance. The Company is pursuing clinical
development with ACI-24.060, an optimized formulation of ACI-24 in
early AD and in population with DS with presence of brain amyloid
pathology.
Mechanism of
Action
|
• |
ACI-24.060’s mechanism of action is similar to the one
described with the original formulation of ACI-24. The
incorporation of T-cell help epitopes in this optimized formulation
is intended to prime, boost and maintain a strong antibody response
against key pathological Abeta species (including oligomeric and
pyroglutamate Abeta). The antibodies elicited by the vaccine in
NHPs showed clear target engagement by binding to human Abeta
plaques on AD patient-derived brain tissue.
|
AC Immune submitted a Clinical
Trial Application (CTA) to the UK Medicines and Healthcare Products
Regulatory Agency (MHRA) in Q4 2021 to initiate development of the
optimized formulation of ACI-24 in patients with prodromal AD and
in adult subjects with DS with presence of brain amyloid pathology.
It then plans for a subsequent Investigational New Drug (IND)
application in the USA in early 2023 for the global development of
the vaccine candidate.
ACI-7104 – anti-a-syn vaccine
Neurodegenerative conditions
with a-syn accumulation, such as Parkinson’s disease, are
increasingly linked to dementia and movement disorders in the aging
population.
ACI-7104 is an optimized
peptide-conjugate vaccine formulation designed to induce
a-syn-specific antibodies recognizing aggregated a-syn species that
have been demonstrated to be toxic to neurons. In contrast,
ACI-7104-induced antibodies do not bind to the monomeric,
physiological form of a-syn and do not cross-react with other
members of the synuclein family such as beta- and
gamma-synuclein.
A substantial package of
preclinical and clinical data has been generated with PD01
(predecessor of ACI-7104). This candidate was tested in two
different transgenic mouse models of PD and Dementia with Lewy
bodies (DLB), the mThy1- and the PDGF-human a-syn transgenic mice.
Active vaccination with PD01 resulted in decreased a-syn pathology
in brain areas most affected by transgene overexpression, including
the substantia nigra and the striatum, and this was accompanied by
a reduced neurodegeneration and by improvements in motor and memory
deficits in both in vivo
models.
PD01 was the first vaccine
candidate against pathological a-syn to be tested in a clinical
study involving patients with early PD. A series of Phase 1 studies
were completed in June 2018. The start of the Phase 2 trial in
early PD patients to evaluate ACI-7104 is scheduled to commence in
H2 2022.
Mechanism of
action
|
• |
ACI-7104 is composed of a short engineered antigenic a-syn
peptide. This peptide coupled to a carrier protein facilitates the
induction of an a-syn-specific antibody response that binds to
toxic aggregated a-syn species with high selectivity (Mandler-M
et al., Acta Neuropathol.
2014).
|
|
• |
Vaccination of wild-type and transgenic mice, resulted in high
antibody titres in plasma, which crossed into the cerebrospinal
fluid (CSF) (Mandler-M et
al., Acta Neuropathol. 2014) and recognized a-syn
aggregates. Vaccination resulted in a decreased aggregation and
accumulation of a-syn oligomers in brains of transgenic animals
(Figure 16).
|
|
• |
Clearance of a-syn was accompanied by reduced
neurodegeneration and by improvements in motor and memory deficits
in both in vivo models
(Mandler-M et al., Acta
Neuropathol. 2014).
|
Figure 16:
Immunization with PD01 reduced the accumulation of a-syn aggregates
in brains of mThy1-human a-syn transgenic mice
Ref: Mandler M et al., Acta Neuropathol. 2014
Clinical
development
Phase 1 study
design
The safety tolerability and
immunogenicity of the a-syn vaccine were studied over a three and a
half year period in 24 early PD subjects. There were four
consecutive studies in this group of patients as shown in Figure 17
(Volc et al., The Lancet
Neurology 2020 Jul), with patients randomized to receive a lower or
higher dose of the alpha synuclein vaccine. After four priming
doses, subjects were re-randomized to receive a booster injection
at one of the two doses, followed by a second booster injection at
the high dose.
Figure 17:
Phase 1 study design
Ref: Volc et al., The Lancet Neurology,
2020
Safety
This Phase 1 study series
demonstrated a favorable long-term safety profile for PD01.
Antibody
response
PD01 induced a long lasting and
boostable antibody response (Figure 18A). Such induced antibodies
have been shown to bind preferentially the aggregated species of
a-syn. The induced antibodies were also demonstrated to bind to
a-syn aggregates in human PD and DLB brain tissue.
Pharmacological
and clinical effect
Evidence
for in vivo target
engagement and signals for clinical efficacy have been observed in
these Phase 1 studies in PD, as immunization was associated with a
decrease in oligomeric a-syn in CSF of treated patients and with a
stabilization of clinical scores as shown by the MDS-UPDRS part 3
scores (Volc et al., The
Lancet Neurology, 2020 Jul). Post hoc analyses of this study series
delivered highly encouraging data with respect to target engagement
and identification of a potential biomarker for PD,
including:
|
• |
In vivo target
engagement of induced antibodies was demonstrated by lowering of
oligomeric a-syn in CSF of vaccinated subjects (Figure 18B).
|
|
• |
A highly significant correlation between oligomeric a-syn
concentration in CSF and MDS-UPDRS 3 score (motor-symptoms) in PD
patients at baseline was shown for the first time.
|
|
• |
The reduction of oligomeric a-syn in CSF correlated
significantly with clinical improvement, the changes in MDS-UPDRS 3
score over time (Figure 18C).
|
Figure 18:
Pharmacokinetic and pharmacodynamic effect of PD01 vaccination in
early PD patients
Ref: Volc et al., The Lancet Neurology,
2020
These combined data provide a
validation of the role of a-syn in disease progression and
demonstrate that a-syn directed vaccination using ACI-7104 has the
potential to positively impact clinical outcome. These data also
provide an excellent basis for design of the planned Phase 2
study.
Semorinemab
Semorinemab is a humanized
high-affinity IgG4 isotype antibody candidate that binds all forms
of Tau. Semorinemab is designed to intercept extracellular Tau,
stopping or slowing cell-to-cell spread and propagation of
pathological Tau in the brain. Semorinemab is in Phase 2 clinical
development for AD as part of an ongoing collaboration, which was
established in 2012, with Genentech.
Lead
characterization
Our anti-Tau monoclonal antibody
program successfully generated multiple humanized antibodies for
potential use as passive immunotherapies, which are highly specific
for pathological forms of Tau found in AD and other Tauopathies.
Results from preclinical studies demonstrated a reduction in
pathological Tau and improvement of long-term spatial memory.
Efficacy studies run in mouse models of AD and other Tauopathies
exhibited dose–response alleviation of Tau pathology with
behavioral improvements.
Figure 19:
Alleviation of Tau pathology in models of AD
Representative images of hippocampal coronal sections from human
Tau-P301L transgenic mice treated with (A) control antibody or (B) semorinemab, and immunostained for
pathological Tau deposits
Ref:
Ayalon et al., Science
Trans. Med. 2021
Clinical
development
A Phase 1 clinical trial involving
75 subjects evaluated the safety, tolerability, pharmacokinetics
and preliminary data on therapeutic activity of semorinemab in
people with AD and in healthy volunteers. This trial was completed
in the second quarter of 2017. Semorinemab was administered at
single doses of up to 16,800 mg to healthy volunteers, and at
multiple doses of 8,400 mg to healthy volunteers and patients with
mild-to-moderate AD. No dose-limiting toxicities and no SAEs were
observed. No participant withdrawals, modifications or
interruptions due to an adverse event were reported. Results were
presented at multiple conferences, including the 13th
International Conference on Alzheimer’s & Parkinson’s Diseases
and Related Neurological Disorders (AD/PD) in 2017, the AAIC in
2017, and the 10th
international CTAD conference in 2017.
Semorinemab exhibited a
dose-proportional pharmacokinetic profile and CNS exposure, with a
median half-life of 32.3 days. Plasma total Tau concentration
increased with increasing drug doses and was doubled in
participants with AD compared with healthy volunteers, suggesting a
pharmacodynamic signal as shown in the figure below.
Figure 20:
Phase 1 pharmacokinetic and plasma Tau results
Ref: Kerchner et al., CTAD 2017.
A Phase 2 clinical trial (Tauriel)
commenced in Q4 2017 with the dosing of the first patient. This
multicenter trial, which enrolled 457 participants, assessed the
safety, tolerability and efficacy of semorinemab in people with
prodromal-to-mild AD. Participants received one of three active
doses or a placebo for 72 weeks, followed by a 96-week optional
open-label extension (OLE) (Figure 21). Primary endpoints included
safety assessment and the composite functional and cognitive
endpoint CDR (Clinical Dementia Rating scale) CDR-SB score.
Figure 21:
Phase 2 (Tauriel) study design
Ref: Kerchner et al., CTAD 2017
On September
23, 2020, the Company reported that Genentech informed us of top
line results which showed that semorinemab did not meet its primary
efficacy endpoint of reducing decline on Clinical Dementia
Rating-Sum of Boxes (CDR-SB) compared to placebo. Two secondary
endpoints, Alzheimer’s Disease Assessment Scale-Cognitive Subscale
13 (ADAS-Cog13) and Alzheimer’s Disease Cooperative Study Group –
Activities of Daily Living Inventory (ADCS-ADL), were also not met.
The primary safety endpoint was however met.
Further analyses revealed a
dose-dependent increase in serum pharmacokinetics and evidence of
target engagement, measured by an increase in plasma Tau levels,
which is consistent with previous Phase 1 study results (Figure
22).
Figure 22:
Phase 2 (Tauriel) study peripheral pharmacokinetic and
pharmacodynamic results
Ref: Teng et al., CTAD 2020
Semorinemab did not show a
dose-dependent effect on Tau PET signal in the brain. However there
was evidence of central target engagement from assessment of
soluble tau in CSF: N-terminal Tau increased with exposure, and
pTau181 and mid-domain Tau decreased.
Figure 23:
Phase 2 (Tauriel) study CSF target engagement
Ref: Wildsmith et al., AD/PD 2021
A second Phase 2 trial (Lauriet)
was initiated in Q1 2019. This is a multicenter study enrolling 272
participants, and is designed to evaluate the clinical efficacy,
safety, pharmacokinetics and pharmacodynamics of semorinemab in
patients with moderate AD [Mini Mental State Examination (MMSE)
16–21, CDR-GS 1 or 2]. The study consists of a screening period, a
double-blind treatment period of 49 weeks, an optional OLE period,
and a follow-up period, with the 11-item Alzheimer’s Disease
Assessment Scale-cognitive subscale (ADAS-Cog11) and Alzheimer’s
Disease Cooperative Study-Activities of Daily Living tools as the
primary endpoints, and CDR-SB, MMSE and safety as secondary
endpoints. Primary completion (last patient, last visit) was in Q2
2021.
On August 31, 2021 the Company
reported that Genentech had informed the Company that the Lauriet
study had met one of its co-primary endpoints, ADAS-Cog 11. The
second co-primary endpoint, ADCS-ADL, was not met. Safety data
showed that semorinemab was well tolerated with an acceptable
safety profile and no unanticipated safety signals. On November 10,
2021, the Company reported that Genentech had presented the full
top-line data from the Lauriet study during a late-breaking session
at the 14th
Clinical Trials on Alzheimer’s Disease conference.
272 subjects were randomized
into the study and 267 dosed. 49 study centers participated in the
US, France, Spain and Poland. In a modified intent to treat (mITT)
population of all trial participants who had received at least one
dose of study drug and had at least one post-baseline ADAS-Cog 11
assessment, there was a 42.2% slowing of cognitive decline compared
to placebo, the result being highly statistically significant
(p=0.0008).
Figure 24:
Effect of semorinemab on cognition assessed by ADAS-Cog11
Ref: Monteiro C. et al., CTAD 2021
The effect on ADAS-Cog 11 was
consistently seen at around the same magnitude in subgroup analyses
looking at subjects with higher or lower baseline severity assessed
by MMSE, brain tau load assessed by GTP1-PET and different ApoE
genotypes. The effect on ADAS-Cog11 was driven by an effect on
memory items in the scale. No changes were apparent in functional
measures including the co-primary endpoint ADCS-ADL, and the
secondary endpoint, CDR-SB, and there was no significant effect in
the other secondary endpoint, the MMSE. The reason for the lack of
functional effects is unclear. Regular interim analyses in the
ongoing extension study are being made to test for later effects on
function. Plasma tau rose markedly during the study confirming
peripheral target engagement, with serum levels of semorinemab in
the expected range. The ratio of CSF to plasma semorinemab
concentration was 0.29%, in the expected range for similar
monoclonal antibodies. There was no apparent effect on global or
regional brain tau load assessed by GTP-1 PET. CSF biomarker data
are not yet available. Safety data indicated that semorinemab was
well tolerated, with no difference in the frequency of serious or
non-serious adverse events or discontinuations due to adverse
events.
The results are the first
evidence for an effect of an anti-tau immunotherapy on cognition.
Data from the ongoing open-label extension phase of the study and
CSF biomarkers when available will help in further interpretation
of the results.
Crenezumab
Crenezumab is a humanized,
conformation-specific monoclonal antibody that targets misfolded
Abeta and has a broad binding profile. Crenezumab was developed
using our proprietary SupraAntigen platform. In 2006, we licensed
crenezumab to Genentech, a company with a long history of
developing and commercializing innovative biologics.
Mechanism of
action
|
• |
Crenezumab binds to multiple forms of Abeta, particularly
oligomeric forms, which it binds to with ten times higher affinity
than to monomers. This is a desirable property since oligomeric
forms of Abeta are believed to be principally responsible for
neurotoxicity in AD.
|
|
• |
Crenezumab localizes to brain regions rich in oligomers,
including the halo around plaques and hippocampal mossy fibers, but
not to vascular Abeta (Maloney et
al., 2019).
|
|
• |
Crenezumab has been designed with an IgG4 backbone to reduce
effector function on microglia compared with an IgG1 backbone, and
to clear Abeta from the brain while limiting inflammation by
minimizing FcγR-mediated inflammatory activation of microglia
(Adolfsson et al., J.
Neurosci 2012).
|
Figure 25:
Crenezumab’s IgG4 backbone balances efficacy with safety
Ref: Data reported in Adolfsson
et al., J. Neurosci
2012
The potential for a better safety
profile derived from a human IgG4 rather than a IgG1 backbone has
been born out in practice by the safety findings from the Phase 1,
2 and 3 clinical studies of crenezumab, in which, following either
single or multiple doses, no increase in ARIA-E was reported
(Cummings et al., 2014 and
Cummings et al.,
2018).
|
• |
Due to its capacity to bind to multiple forms of Abeta, with
10-fold higher specificity to oligomers, which are thought to be
the most toxic species, crenezumab also protects against
oligomer-induced neurotoxicity.
|
|
• |
Linked to its unique epitope, crenezumab has been shown to
promote disaggregation of existing Abeta aggregates and to disrupt
their assembly, preventing amyloid plaque formation. The crystal
structure reveals binding interactions that are consistent with
this flexible binding profile and provides further explanation for
crenezumab’s ability to block aggregation and to promote
disaggregation.
|
Signal of
activity in patients with milder AD (MMSE 22–26) in Phase 2
clinical trials
|
• |
In the proof-of-concept Phase 2 studies of crenezumab, a
positive trend in cognition was observed, with a greater effect on
cognition in patients with a milder stage of AD (MMSE 22–26).
|
|
• |
In the ABBY cognition study, there, was a statistically
significant 35% reduction in the rate of cognitive decline in the
non-pre-specified milder AD patient population (MMSE 22–26) for the
high-dose arm.
|
|
• |
In the BLAZE biomarker study, the high-dose arm showed a
consistent trend of reduced Abeta accumulation in the brain over
time, as shown in two independent exploratory analyses of
florbetapir-PET data. In addition, results have shown that
crenezumab has the ability to enhance the removal of these proteins
from the brain as evidenced by a significant increase in CSF Abeta,
confirming target engagement by crenezumab.
|
Favorable safety
profile allowing for higher dosing
|
• |
Phase 2 data from ABBY and BLAZE studies suggested that there
were no imbalances in overall rate of AEs, and these were not
dose-related, with only one case of asymptomatic ARIA-E (0.4% in
ABBY, 0.3% on active pooled) in patients treated with crenezumab.
AEs also included inflammation of the throat and nasal passages,
urinary tract infections and upper respiratory infections. However,
no patients in the studies experienced SAEs that were believed
related to the administration of crenezumab.
|
|
• |
A Phase 1 study with higher doses of crenezumab up to 120
mg/kg showed good tolerability with no investigator assessed
drug-related SAEs and no events of ARIA-E, supporting the dose of
60 mg/kg in the Phase 3 CREAD clinical trials.
|
|
• |
The good safety profile and lack of induction of ARIA-E was
confirmed in the Phase 3 CREAD and CREAD 2 studies, in which there
was no increase in incidence of SAEs compared with placebo.
|
|
• |
Crenezumab is currently being evaluated in a Phase 2 clinical
prevention trial in Colombia, which has enrolled 300 cognitively
healthy individuals of whom 200 are genetically predisposed to
develop early AD. As of January 2019, two Phase 3 clinical trials,
CREAD and CREAD 2, in patients with prodromal-to-mild AD were
discontinued after an interim analysis of the CREAD study conducted
by our collaboration partner Genentech.
|
Clinical development
Phase 2
studies
Phase 2 study
design overview
Crenezumab has been studied in two
Phase 2 clinical studies, the ABBY proof-of-concept study and the
BLAZE biomarker study. These two studies enrolled a total of 522
patients. The purpose of these studies was to investigate whether
crenezumab could delay cognitive and functional decline and reduce
the accumulation of brain amyloid in patients with mild-to-moderate
AD. The sample size of the studies was not expected to have
adequate power to detect a modest but clinically significant
difference between active medication and placebo at the 5%
significance level (as is commonly the case in Phase 2 studies in
AD). Instead, consistent trends across different endpoints and dose
dependencies were considered indicators of a response in this
learning phase of development, with confirmation to then be sought
in Phase 3. Both studies had two active arms: a low-dose arm
receiving 300 mg subcutaneous injection, every 2 weeks and a
higher-dose arm receiving 15 mg/kg intravenously every 4 weeks. The
primary analysis was conducted at 73 weeks, after 68 weeks of
treatment. Safety and tolerability measures included repeated MRI
scans to assess for the development of ARIA, both vasogenic edema
(E) and hemorrhages (H).
ABBY study
results
In the ABBY study, a positive trend
in cognition was observed with a greater effect on cognition in
patients with a milder stage of AD (MMSE 22–26), although the study
did not meet its co-primary endpoints in patients with
mild-to-moderate AD (MMSE 18–26). There was no significant change
in cognition in patients who received low-dose subcutaneous
crenezumab. Results of an exploratory analysis of the high-dose
intravenous arm demonstrated that patients with the mildest
cognitive impairment at screening (MMSE 22–26) showed a
statistically significant 35% slowing of the rate of cognitive
decline over 73 weeks. The effect became greater over time, as
shown by the increasing separation of the crenezumab (solid line)
and placebo (dashed line) curves in the figure below. The milder
group was not pre-specified, meaning the group of patients with
milder AD was not identified before commencing the Phase 2 clinical
studies.
Figure 26: ABBY
high-dose arm: Change in ADAS-Cog 12
Ref: Cummings et al., AAIC, 2014
An exploratory subanalysis in a
non-pre-specified subgroup of patients with milder symptoms (MMSE
22–26) showed a 35.4% reduction in cognitive decline. The sample
size of the study was not expected to have adequate power to detect
a modest but clinically significant difference between active
medication and placebo at the 5% significance level (as is commonly
the case in Phase 2 studies in AD). Instead, consistent trends
across different endpoints and dose dependency are considered
indicators of a response in this learning phase of development,
with confirmation then sought in Phase 3. In the pre-specified
subgroup analysis in patients with mild AD (MMSE 20–26), treatment
with high-dose intravenous crenezumab led to a 23.8% reduction in
cognitive decline. In patients with mild-to-moderate AD (MMSE
18–26) treated with high-dose intravenous crenezumab, there was a
16.8% reduction in cognitive decline. Effect sizes and p-values for
exploratory analyses were not adjusted for multiplicity.
BLAZE study
design
The BLAZE study was a randomized,
double-blind, parallel-group, placebo-controlled study to evaluate
the effects of crenezumab on brain amyloid burden as assessed by
amyloid PET imaging and other biomarker endpoints in patients with
mild-to-moderate AD. The primary endpoint was the change in brain
amyloid load using florbetapir-PET. The terms “brain amyloid
burden” and “brain amyloid load” refer to the total amount of
amyloid deposited in the brain. In total, 91 patients were included
in the study.
BLAZE study
results
The primary endpoint of change in
brain amyloid load by florbetapir-PET was not met, but the study
was not powered to detect statistically significant results.
However, positive trends were observed as shown below in
exploratory analyses of the BLAZE amyloid PET results using a white
matter reference region, which is considered a more sensitive
approach for longitudinal studies. These analyses, conducted
independently by two laboratories, the Banner Alzheimer’s Institute
and MNI Laboratories, produced analogous results, with a trend in
the reduction of Abeta accumulation observed in the high-dose arm
(Figure 27). As described below, a similar result was obtained in
the Phase 3 studies.
Figure 27:
Blaze high-dose arm: amyloid PET results
Ref: Honigberg et al., CTAD 2014
The BLAZE biomarker study high-dose
intravenous cohort showed a consistent trend of reduced Abeta
accumulation in the brain over time as shown by two independent
exploratory analyses of florbetapir-PET data. Using white matter
rather than cerebellum as the key reference region in the brain is
generally considered a more robust method of showing treatment
effects of AD therapies.
In the BLAZE study, patients also
showed a statistically significant increase in CSF Abeta1–42,
which we believe confirms target engagement by crenezumab. Similar
results were observed in the ABBY study, which assessed CSF
Abeta1-42
level in 49 patients. These results suggest that Abeta is being
eliminated from the brain when treated with crenezumab.
Figure 28:
BLAZE high-dose arm: crenezumab increases CSF total Abeta levels
relative to placebo
Ref:
Honigberg et al., CTAD
2014
The BLAZE study results suggest
that Abeta is being eliminated from the brain as patients showed a
statistically significant increase in CSF Abeta1–42,
which confirms target engagement by crenezumab.
Safety data
from ABBY and BLAZE studies
Crenezumab demonstrated favorable
safety and tolerability in Phase 2 clinical studies even at high
doses. Crenezumab’s safety profile is especially reflected in a low
incidence of ARIA-E (0.3%) in Phase 2 clinical studies. ARIA-E was
observed in only one patient who received high-dose intravenous
crenezumab in the ABBY study. No case of ARIA-E was reported in the
placebo arm or the BLAZE study. Favorable pharmacokinetic
properties coupled with a favorable safety and tolerability profile
enables crenezumab to penetrate the brain more readily at
therapeutically relevant doses. As dose-limiting toxicities are a
potential reason for the failure of other antibodies to demonstrate
efficacy, crenezumab’s potential safety at high doses is a
distinguishing product feature.
At AAIC in 2014, it was reported
that in the combined Phase 2 study populations, SAEs occurred at
similar rates in patients treated with crenezumab (16.5%) and in
patients given a placebo (11.9%).
Phase 1b study to explore higher
doses
To explore safety at higher doses,
crenezumab was tested in a Phase 1b dose-escalation clinical study
(NCT02353598) conducted in the US. This randomized,
placebo-controlled, double-blind, four parallel-arm study evaluated
the safety and tolerability of at least four doses of intravenous
crenezumab in 77 patients with mild-to-moderate AD (MMSE 18–28)
between the ages of 50 and 90 years. An optional OLE stage was
offered to patients after completion of the double-blind stage of
the study. At the 2017 AAIC meeting, Genentech presented the
results of the four cohorts with mild-to-moderate AD. No
dose-limiting toxicities were observed at crenezumab doses of 30,
45, 60 and 120 mg/kg. No events of ARIA-E were observed and only
few patients (6 of 75) showed asymptomatic ARIA-H. The
pharmacokinetic profile of crenezumab was dose-proportional up to
the 120 mg/kg dose, with the 60mg/kg dose being selected for the
Phase 3 CREAD and CREAD 2 studies.
Phase 2 AD prevention study
There is increasing understanding
from studies in patients at risk of AD due to genetic mutations
that the build-up of Abeta in the brain is a very early event in
the condition, starting around 25 years before symptoms develop
(McDade et al. 2018). To
treat the underlying amyloid pathology effectively it may therefore
be necessary to use anti-amyloid therapies in a preventive mode,
starting in patients in whom symptoms have not yet emerged.
In 2012, crenezumab was
independently selected from among 25 product candidates for use in
the first-ever such AD prevention study. The study, a collaboration
worth USD 100 million between the NIH, Banner Alzheimer’s Institute
and Genentech, is the cornerstone of the global Alzheimer’s
Prevention Initiative. Crenezumab is being administered
pre-symptomatically to 300 members of an extended Colombian family,
of which 200 members carry a mutation that causes early-onset AD.
Family members usually develop symptoms before the age of 45 years.
The 5-year study has cognitive endpoints. An interim analysis is
possible according to the protocol, but the data and results of
that analysis may not be made public due to patient sensitivity.
The study commenced Q4 2013 and the data for the primary outcome
measures are expected in H1 2022.
Figure 29:
Crenezumab AD prevention trial (Alzheimer’s Prevention Initiative
ADAD): unique population to study prevention treatment
(1) Mild cognitive impairment;
(2) Alzheimer’s disease; (3) Presenilin-1
Ref: McDade et al., Neurology 2018
Phase 3 studies (CREAD and CREAD
2)
The randomized, double-blind,
placebo-controlled, parallel-group Phase 3 CREAD study enrolled
about 750 participants with prodromal or mild AD at the age of
50−85 years. A high dose of crenezumab (60 mg/kg) was administered
intravenously once every 4 weeks for 100 weeks. The primary outcome
measure was the change from baseline to week 105 in CDR-SB score.
An exposure–response model to evaluate the best dose of crenezumab
for the treatment of AD was established, which predicted an
improved outcome of the Phase 3 CREAD study by using the higher
dose of 60 mg/kg relative to the Phase 2 trials (Polhamus
et al., CTAD 2016).
On January 30, 2019, we announced
that Roche, the parent company of our collaboration partner, is
discontinuing the CREAD and CREAD 2 (BN29552 and BN29553) Phase 3
studies of crenezumab in people with prodromal-to-mild sporadic AD.
The decision came after an interim analysis of the first CREAD
study conducted by the IDMC, which indicated that crenezumab was
unlikely to meet its primary endpoint of change from baseline in
CDR-SB score.
As presented at CTAD 2019, target
engagement was observed with increases in levels of
Abeta1–42
in blood and CSF. As shown in Figure 30, the number of subjects
available for analysis fell significantly after the baseline. Due
to the early termination of the studies, data at the 2-year
timepoint was only available for 17 of the 139 crenezumab subjects
in whom CSF Abeta was analyzed.
Figure 30: CSF
total Abeta42 and total Abeta change from baseline, pooled
CREAD/CREAD2 results
Ref: Bittner et al., Roche CTAD 2019
Reduced accumulation of Abeta in
the brain on florebetapir amyloid PET scans was observed, with a
pattern very similar to that observed in the Phase 2 BLAZE
studies.
Figure 31:
[18F] Florbetapir amyloid PET standard uptake value ratio (SUVR)
change from baseline, pooled CREAD/CREAD2
Ref: Bittner et al., Roche CTAD 2019
A numerical trend to reduction in
level of total Tau and phospho-Tau 181 (pTau181) in the CSF in
patients on crenezumab compared with placebo was observed although
the small numbers in the analysis due to early termination of the
studies preclude firm conclusions from being drawn.
Figure 32: CFS
total Tau and pTau181 change from baseline, polled
CREAD/CREAD2
Ref: Bittner et al., Roche CTAD 2019
Positive trends on a range of
biomarkers associated with AD in CSF including neurogranin,
neurofilament light chain (NFL), Glial fibrillary acidic protein
(GFAP), soluble Triggering receptor expressed on myeloid cells 2
(sTREM2), Chitinase-3-like protein 1(YKL-40) and a-syn were
reported by Roche at the CTAD 2019 conference, although again the
small numbers due to early termination of the studies limit
interpretability of the results.
Figure 33:
Exploratory biomarkers: Roche NeuroToolkit
Ref: Bittner et al., Roche CTAD 2019
Safety in the
CREAD and CREAD 2 studies
The decision to terminate the CREAD
and CREAD 2 was not related to safety. No safety signals for
crenezumab were observed in this analysis and the overall safety
profile was similar to that seen in previous trials. There was no
difference in the rate of newly developing ARIA-E (0.3%) between
the active and placebo arms and the rates of ARIA-H were also
similar (8.8% on crenezumab vs 6.8% on placebo).
Prevention
trial in familial AD
As described above crenezumab
continues to be studied under the Alzheimer’s Prevention Initiative
in a preventive trial in Colombia, which began in 2013, of
cognitively healthy individuals with an autosomal-dominant mutation
who are at risk of developing familial AD.
Morphomer Tau
Approximately 2,000 compounds were
screened so far for the Morphomer Tau program. This allowed the
identification of several chemical series of orally bioavailable
small molecules with suitable CNS properties. The lead compounds
displayed selectivity for binding to pathological Tau aggregates in
preference to other protein aggregates. In addition, the lead
compounds were able to prevent Tau aggregation and promote its
disaggregation. Further characterization using multiple orthogonal
in vitro, ex vivo and in vivo tests addressing pharmacology
absorption, distribution, metabolism, and excretion (ADME), and
safety properties led to the identification of the first clinical
candidate ACI-3024.
ACI-3024
ACI-3024 is a potent Tau
aggregation inhibitor active against the 3R and 4R human Tau
isoforms as well as the mutant forms associated with human
Tauopathies, such as FTLD-Tau (e.g., PSP, Pick’s disease,
corticobasal degeneration). ACI-3024 selectively binds to
aggregated Tau and does not bind to the monomeric forms of Tau.
Moreover, the binding to Tau aggregates is selective, with no
cross-reactivity to aggregates of Abeta and a-syn.
ACI-3024 showed a potent and
dose-dependent reduction in spontaneous intracellular Tau
aggregation and misfolding as measured by immunocytochemistry in
human neuronal-like cells over-expressing Tau. Furthermore,
ACI-3024 promoted ex vivo
disaggregation of Tau neurofibrillary tangles on human AD brain
sections.
The in vivo efficacy of ACI-3024 was
evaluated in the Tg4510 mouse model (Ramsden et al., 2005). In vivo treatment of Tg4510 transgenic
mice with ACI-3024 reduced aggregated and insoluble
hyper-phosphorylated Tau. Immunohistochemistry analysis of
misfolded Tau using an MC1 antibody in Tg4510 brain sections of the
same mice treated with ACI-3024 showed reduction of misfolded Tau.
These effects were proportional to the plasma concentration of
ACI-3024 (Figure 34).
Total Tau concentration in
cerebrospinal fluid (CSF) was inversely correlated with ACI-3024
exposure in plasma, suggesting the possibility of exploring CSF Tau
concentrations as a biomarker of target engagement.
Further work is ongoing to evaluate
ACI-3024 in selected NeuroOrphan indications.
Figure 34:
Dose-dependent reduction in Tau misfolding in vivo
Ref: AC Immune unpublished
data
Preclinical safety
ACI-3024 has a good in vitro and in vivo ADME profile, including low
clearance, long half-life and good CNS disposition as assessed by
brain and CSF concentrations. ACI-3024 was negative in in vitro and in vivo genotoxicity assays [Ames,
micronucleus test (MNT) and mouse lymphoma cell mutagenesis (MLY)]
and has undergone an extensive toxicology and safety pharmacology
assessment. The no observed adverse effect level has been
established at 300 mg/kg in rodent and at 450 mg/kg in non-rodent
animals after 4 weeks of treatment (Poli, CTAD 2018).
Effect on neuroinflammation
ACI-3024 efficacy on pathological
Tau-induced neuroinflammation was assessed in vitro and in vivo. In vitro, ACI-3024 induced a potent
reduction of Tau-induced neuroinflammation markers (Figure 35,
below). In vivo, in Tg4510
mice, treatment with ACI-3024 overall reduced microgliosis, most
likely as a downstream consequence of reducing Tau pathology, by
reducing the derived pathological Tau-induced microglial activation
(Figure 35).
Figure 35:
ACI-3024 significantly reduces Tau-induced neuroinflammation
Ref: AC Immune unpublished
data
Clinical development
Phase
1 study
This Phase 1 study was a
first-in-human (FiH), randomized, placebo-controlled, double-blind,
sequential single and multiple ascending dose study. The study
assessed the safety, tolerability, pharmacokinetics, and
pharmacodynamics of ACI-3024. Part I included five single ascending
doses in healthy volunteers, with a food effect assessment in the
fourth dose cohort. In Part II, three escalating multiple dose
regimens were evaluated; regimen two was assessed in different
populations of healthy volunteers. CSF samples were collected from
the highest multiple dose group.
The study was executed as
planned and all single and multiple dosing regimens were completed
in healthy young, elderly, and Japanese subjects. ACI-3024 was
administered following single or multiple oral doses and
dose-dependent plasma exposure was observed. ACI-3024 showed a long
half-life (47.5 to 101 h), with steady state reached after 12-13
days. Low renal clearance was shown. After multiple doses, ACI-3024
concentrations in CSF exceeded target concentrations based on
animal studies.
Plans to conduct additional
clinical trials with ACI-3024 in AD have been suspended. The
Companies have decided to pursue other promising Tau Morphomer
candidates from AC Immune’s research platform for potential
clinical development in AD.
Figure
36: Morphomer Tau therapeutic program: summary and outlook
Tau diagnostics
The severity of cognitive
impairment in patients with AD is correlated with the presence of
Tau protein tangles, leading us to believe that an imaging agent
for Tau is equally, if not more important than Abeta-PET to assess
spreading of pathology in the brain. In May 2020, Eli Lilly
received FDA approval for the first Tau PET tracer TAUVID
(flortaucipir F18 injection). However, TAUVID received approval
only for a pathology indication (i.e., correlation with
histopathology findings in Braak 5 and 6 patients), but has not
received a prognostic label (i.e., prediction of cognitive
deterioration based on a positive Tau PET scan.)
Our Tau-PET tracers are designed to
bind specifically to the pathological forms of human Tau in AD and
other Tauopathies. They have demonstrated an excellent PET tracer
profile with their ability to cross the blood brain barrier and a
high selectivity to pathological Tau even in the early-stage
disease.
In May 2014, we established a
license and collaboration agreement for our Tau-PET imaging program
with LMI. The Phase 1 clinical study of our clinical candidate
PI-2620 in AD was completed in Q1 2018. LMI commenced a Phase 2
longitudinal study in AD of the program in Q3 2019. The Phase 2
longitudinal study in AD in South Korea (Asan Medical Center) was
completed in Q4 2021.
PI-2620 is selective for Tau over
Abeta and other “off-target” binding compared with current
published Tau-PET agents in development, as no binding to Abeta
in vivo and no
“off-target” retention in basal ganglia or choroid plexus was
observed. In addition, PI-2620 can be readily radiolabeled with
fluorine 18. A major differentiator for PI-2620 is its ability to
bind 4-repeat (4R) Tau isoforms, which are present in varying
amounts in different neurodegenerative diseases. Most Tau PET
tracers in development are not able to bind 4R Tau and are of
limited use for certain diseases driven by these Tau species. Thus,
two test-retest studies (Phase 1) in PSP are open and recruiting
with results anticipated in H2 2022. Due to its ability to detect
4R Tau aggregates, PI-2620 is the first and only PET imaging agent
to receive orphan drug designations for the diagnosis of PSP and
CBD.
Figure 37:
PI-2620 – Distinguishing the clinically predicted Tau isoform in
different tauopathies
Ref: Song et al., J Cereb Blood Flow Metab.
2021
Figure 37 above shows Abeta PET
images (left) together with Tau PET (right) SUVR images of cortical
areas of patients with 3/4R (top), 4R (middle) tauopathies and
healthy control (bottom). Thus, PI-2620 binding characteristics in
cortical regions differentiated between 3/4R- (AD) and
4R-tauopathies (PSP, CBD) and might serve as a supportive readout
in the diagnostic workup of neurodegenerative disorders.
Figure 38:
PI-2620 – differentiated CBD subtypes
Ref: Palleis et al., Mov Disord. 2021
The PI-2620 Tau-PET data displayed
in Figure 38 above shows the average PI-2620 distribution volume
ratio (DVR) binding maps presented as axial overlays on a standard
MRI template for all study groups (Aβ[-]CBS, n = 34 (top);
Aβ[+]CBS, n = 11 (middle); and controls (CTRL), n = 14 (bottom)).
Extracerebral voxels were masked. Images from patients with
left-dominant symptoms were flipped. Thus, the data indicated a
value of PI-2620 for evaluating corticobasal syndrome, providing
quantitatively and regionally distinct signals in
β-amyloid-positive as well as β-amyloid-negative corticobasal
syndrome. In corticobasal syndrome, PI-2620 may potentially serve
for a differential diagnosis and for monitoring disease
progression.
Figure 39:
PI-2620 – in silico
docking of PI-2620 in AD
Ref: Kroth et al., J Med Chem. 2021
Figure 39 above shows the
in silico docking of
PI-2620 into site1 utilizing a dodecamer model of paired helical
filament (PHF) (A). Potential binding interactions between PI-2620
and adjacent amino acid residues of site1 are indicated with black
arrows (B).
Tau diagnostics are a major market
opportunity that will be driven by the growth in the aging
population and the testing and availability of disease-modifying
drugs. We believe a best-in-class Tau tracer has the potential to
achieve a substantial global market share in this large and growing
market, which includes AD as well as other important
Tauopathies.
A-syn diagnostics
We are also developing PET imaging
agents to detect a-syn, which progressively accumulates in the
brains of PD patients and is believed to be central to the
neurodegenerative process of PD, as well as several other
disorders, including Lewy body dementia and MSA, making it a
priority target for development of therapeutics and diagnostics. We
have identified molecules leveraging our Morphomer technology that
selectively bind to a-syn pathological structures from human PD
brain with affinity in the low-nanomolar range.
In 2021, we completed the clinical
evaluation of our second generation clinical candidate ACI-3847 by
showing no differences in tracer retention between in patients with
synucleinopathies compared to controls.
In the same timeframe, we have also
initiated the clinical development of our third-generation
candidate, ACI-12589. Compared to the second-generation tracer,
ACI-12589 retained the good selectivity and pharmacokinetic profile
while also showing a significantly increased signal specificity in
PD versus non-diseased human tissues (Figure 40). The current
status of the program was presented at AD/PD 2021 and AAIC
2021.
The Phase 1-enabling
preclinical and manufacturing activities for ACI-12589 were
completed in 2020 and the IND was accepted in early 2021. The FiH
clinical evaluation of the third-generation tracer in PD, MSA and
other a-synucleinopathies began in February 2021 and the readout is
expected by Q2 2022.
Figure 40:
3rd
generation alpha-synuclein tracers with improved properties
(1) Parkinson’s disease; (2)
Alpha synuclein; (3) Parkinson’s disease with dementia; (4) PD with
SNCA G51D mutation; (5) Non-diseased control; (6)
Non-specific
Ref: AC Immune, AAIC Conference,
2020
Currently there are no imaging
products in the market that target a-syn. This provides us with the
opportunity to become the market leader in a-syn PET imaging. We
believe the ability to image a-syn deposits in the brain will
enable a fundamental change in the approach toward diagnosing and
treating PD and other a-syn-associated diseases.
Our preclinical programs
Using our SupraAntigen and
Morphomer platforms, we have generated additional discovery and
preclinical stage molecules targeting key pathologies that drive a
range of neurodegenerative diseases, including TDP-43, a-syn, and
NLRP3. We are accelerating the development of several therapeutic
product candidates currently in preclinical development, including
several programs focused on indications outside of AD as a critical
part of our expansion strategy.
Figure 41: Key
pathologies for further pipeline expansion
(1) Frontotemporal lobar
degeneration; (2) Amyotrophic lateral sclerosis; (3)
Limbic-predominant age-related TDP-43 encephalopathy; (4) TAR
DNA-binding protein 43; (5) Alpha-synuclein; (6) NOD-like receptor
protein 3
Based on the data to date, our
technology platforms can be applied to misfolded proteins across a
broad range of indications. Five of our preclinical programs are
outlined below:
Product
candidate
|
Target
|
Lead
application
|
Partner
|
Platform
|
a-syn antibody
|
a-syn
|
PD, NeuroOrphan
|
Proprietary
|
SupraAntigen
|
Morphomer a-syn
|
a-syn
|
PD, NeuroOrphan
|
Proprietary
|
Morphomer
|
Anti-TDP-43
antibody
|
TDP-43
|
NeuroOrphan
|
Proprietary
|
SupraAntigen
|
Morphomer
inflammasome
|
NLRP3-ASC
|
CNS, non-CNS
|
Proprietary
|
Morphomer
|
Anti-inflammasome
antibody
|
NLRP3-ASC
|
CNS
|
Proprietary
|
SupraAntigen
|
AC Immune’s proprietary
SupraAntigen platform is used to generate antibodies that can be
used as therapeutic and diagnostic products. Such antibodies are
generated by injecting the full-length protein and/or corresponding
peptide constructs in mice and by selecting the antibodies for
their ability to bind to and break up aggregated forms of misfolded
proteins. The a-syn and TDP-43 antibodies were discovered using the
SupraAntigen technology platform. Both antibodies have unique
binding properties allowing them to bind to unique epitopes of the
pathological forms of a-syn and TDP-43, respectively.
A-syn antibody
The a-syn antibodies generated
using our SupraAntigen platform have unique binding properties
allowing them to bind preferentially to the pathological forms of
a-syn. A-syn aggregation and spreading are established targets for
PD, MSA and other synucleinopathies. Antibodies that interfere with
the aggregation and spreading mechanisms of a-syn provide a
therapeutic option for the treatment of PD. The a-syn antibodies
were able to significantly delay the seeded aggregation of
pathological a-syn in an in
vitro aggregation assay, and were able to significantly
decrease pathological a-syn spreading in an in vivo animal model of PD.
Characterization using multiple orthogonal in vitro and in vivo tests addressing binding,
specificity, functionality and pharmacological properties has led
to the identification of the lead candidate ACI-5755.
Lead
characterization
ACI-5755 selectively binds to
pathological forms of a-syn with low-nanomolar affinity and shows a
significant preference over monomeric a-syn. Additionally, ACI-5755
shows strong recognition for pathological a-syn in patient-derived
tissues in both PD and MSA. ACI-5755 showed a potent and
dose-dependent reduction in the seeding capacity of pathological
a-syn in a proprietary in
vitro aggregation assay. Moreover, ACI-5755 substantially
reduced the propagation of a-syn aggregates in a cell-based model.
The in vivo efficacy of
ACI-5755 was evaluated in the M83 propagation mouse model (Luk
et al., 2012). Treatment
of mice with ACI-5755 significantly decreased pathological a-syn
spreading in vivo.
Furthermore, a significant reduction in the rate of body weight
loss compared with the vehicle-treated control group was observed
for mice treated with ACI-5755.
Morphomer a-Syn
Leveraging our Morphomer
platform, we identified and characterized the first biologically
active small molecule inhibitors targeting intracellular a-syn
aggregates. Initial compounds, from several distinct chemical
series, significantly decrease a-syn aggregate accumulation in
neurons by interfering with the fibrillation process. Iterative
medicinal chemistry optimization led to the identification of
orally available and well-tolerated compounds with favorable
CNS-penetrant pharmacokinetic properties, which will be progressed
into in vivo
proof-of-concept studies in animal models of
alpha-synucleinopathies. Medicinal chemistry efforts will continue
on improving properties of lead chemical series, in parallel to
identifying structurally diverse compounds fulfilling the target
product profile.
In December 2021, the Company
received a grant from the MJFF to fund the optimization of the
current compound series and declare a preclinical lead, which will
commence in Q1 2022.
TDP-43 antibody
TDP-43 is a recently identified
target of growing interest for NeuroOrphan indications such as
frontotemporal dementia (FTD) and ALS. Interestingly, TDP-43 also
plays an important role in other significant neurodegenerative
indications such as AD or LATE.
Anti-TDP-43 antibodies binding to
various regions of TDP-43 were generated by our SupraAntigen
platform. A subset displayed conformational selectivity to
misfolded TDP-43, while others recognized all TDP-43 isoforms
(Figure 42A). Multiple antibodies were generated and characterized
in vitro, from which two
pan-TDP-43 antibodies (ACI-5891 and ACI-5886) were selected for the
evaluation of their efficacy in mitigating TDP-43 aggregation
in vitro and in vivo (Figure 42B-C). ACI-5891
showed a high binding affinity for TDP-43 and ability to reduce
TDP-43 aggregation in vitro and
in vivo.
Lead
characterization
To evaluate the functional
efficacy of TDP-43 antibodies in
vitro, the ability of ACI-5891 to inhibit TDP-43 aggregation
was tested. In an in vitro
assay with recombinant TDP-43, ACI-5891 significantly inhibited
TDP-43 aggregation by 98% compared with the isotype control and
significantly promoted their phagocytosis by mouse primary
microglia. Using FTLD-TDP patient-derived brain extracts to induce
templated TDP-43 aggregation in
vitro, ACI-5891, which binds to the C-terminal domain of
TDP-43, was able to substantially interfere with this process of
seeding (Figure 42YB). Moreover, ACI-5891 demonstrated functional
efficacy in vivo by
reducing pathological TDP-43 in two different mouse models of ALS
and FTD (Figure 42C). Our findings demonstrate, for the first time,
that a monoclonal antibody targeting the C-terminal region of
TDP-43 limits pathology and neurotoxicity by enabling clearance of
misfolded TDP-43 through microglia engagement and support the
clinical strategy to target TDP-43 by passive immunotherapy.
ACI-5891
humanization and manufacturability assessment
ACI-5891 was successfully
humanized on a human antibody framework. Several variants had a
similar binding capacity for TDP-43 as well as potency for
inhibition of TDP-43 aggregation as compared to the chimeric
monoclonal antibody. The target values were achieved for the lead
candidates in terms of target affinity, functional efficacy and
percentage humanness. Developability of clinical lead candidates
was further confirmed in manufacturability assessment
studies.
Figure 42: Key
results for TDP-43 antibodies program
Ref: AC Immune, AD/PD
Conference, 2022
Neuroinflammation and the NLRP3
inflammasome pathway
Microglial cells are the main
resident immune cells in the brain, which maintain a healthy
environment by removing damaged cells and misfolded protein
aggregates. When overstimulated, microglia can drive
neuroinflammation, leading to increased neuronal death and disease
progression. A key molecular pathway that is activated by misfolded
proteins related to neurodegenerative and other diseases, is the
NLRP3 inflammasome, a multi-protein complex that forms within
microglia leading to production of pro-inflammatory factors that
exacerbate neuronal atrophy. A critical component of the NLRP3
pathway is ASC (apoptosis-associated speck-like protein containing
a C-terminal caspase recruitment domain), which is formed and
released by activated microglia. Intracellularly, ASC specks
participate in the production of pro-inflammatory cytokines, whereas
extracellular ASC specks cause acute inflammatory reactions. ASC
specks have been identified in microglia within the CNS of patients
with NDD (Venegas, 2017) as well as patients’ body
fluids.
As illustrated in Figure 43,
pathological species of Abeta, Tau, a-syn and TDP-43 induce NLRP3
inflammasome activation and ASC speck formation. AC Immune is
developing multiple small molecule and antibody-based candidates
with the potential to inhibit the NLRP3 pathway. Recent
in vitro studies and
in vivo experiments in
animal models of AD, PD and ALS have validated this approach.
Figure 43:
Proteinopathies exacerbate NLRP3-driven neuronal damage and
promotes further neurodegeneration
(1) Amyotrophic lateral
sclerosis; (2) Frontotemporal lobar dementia; (3) TAR DNA-binding
protein 43; (4) NOD-like receptor protein 3; (5)
Apoptosis-associated speck-like protein containing a CARD, also
PYCARD; (6) Neurofibrillary tangle
Targeting NLRP3-ASC in
neurodegenerative diseases
In AD, Abeta peptides, which
accumulate to form the characteristic plaques in AD activate the
NLRP3 inflammasome (Halle, 2008). The downstream pro-inflammatory
factors, IL-1b and IL-18, increased in cells isolated in these
patients (Saresella, 2016). Further validation of these targets in
AD involve crossing NLRP3 or ASC knockout mice to models of
Abeta-driven pathology. In these models, neuroinflammation
decreased and neuronal and memory function improved (Heneka, 2013;
Demspey, 2017; Venegas, 2017). Recently, ASC speck and IL-18 levels
were shown to be higher in human Mild Cognitive Impairment (MCI)
and AD brain samples indicating that ASC is a promising biomarker
of MCI and AD (Scott, 2020).
In Parkinson’s disease, NLRP3 is
activated and ASC formation is upregulated (Gordon, 2018 and
Anderson, 2021). Exome sequencing analysis identified multiple
single-nucleotide polymorphisms of NLRP3 including rs7525979, which
was associated with a significantly reduced risk of developing PD
(von Herrmann, 2018). In vitro, NLRP3 inhibition decreases
a-syn-mediated inflammasome activation in mouse microglial cells
and extracellular ASC release. In multiple PD animal models,
targeting NLRP3 effectively mitigates motor deficits, nigrostriatal
dopaminergic degeneration and accumulation of a-syn aggregates
(Gordon 2018). Taken together, NLRP3 is responsible for driving
neuroinflammation that results in progressive dopaminergic
neuropathology, highlighting NLRP3 as a potential target for PD
disease-modifying treatments.
As illustrated in Figure 43,
extracellular Tau activates NLRP3 and ASC formation in microglia.
In patients with frontotemporal dementia, elevated cleavage of
caspase-1, increased ASC levels and mature IL-1b are observed
(Ising, 2019). Furthermore, in preclinical models, injection of
fibrillar Abeta induces Tau pathology in an NLRP3-dependent manner
(Ising, 2019) and the absence of ASC or inhibition of NLRP3
decreases seeding by Tau in
vivo and in vitro
(Stancu, 2019). Finally, NLRP3 inhibition ameliorates inflammation
and ER stress signaling in a model of Tau-driven pathology, as well
as partially normalizes phospho-tau levels (Hull, 2020).
Concerning ALS, TDP-43-mediated
activation of microglia causes motor neuron cell death in vitro (Zhao, 2015) with downstream
activation of NF-kB and NLRP3 (Clark, 2020) and involves CD14. This
finding is clinically relevant as increased CD14 expression by
microglia is observed in postmortem spinal cord tissue from
patients with ALS, a TDP-43-driven disease (Clark, 2020).
Furthermore, wildtype and mutant forms of TDP-43 activate microglia
to generate IL-1b, which is abolished by NLRP3 inhibition (Deora,
2019).
Microglia isolated from the ALS
mouse model, SOD1G93A, express elevated levels of NLRP3 (Deora,
2019). When microglia are incubated with soluble or aggregated
SOD1G93A, NLRP3 is activated, ASC specks are formed and IL-1b is
secreted which is prevented by treatment with an NLRP3 inhibitor
(Deora, 2019).
Our strategy for targeting the
NLRP3-ASC inflammasome
AC Immune is aggressively pursuing
this key pathway in order to reduce the unwanted progression of
inflammation in diseases and syndromes caused by the
hyper-activation of the NLRP3 inflammasome. Our aim is to develop
therapeutics that decrease production of pro-inflammatory factors
yet maintain normal phagocytosis of debris and misfolded proteins
as well as allow the function of other pathogen-sensing pathways.
Currently, AC Immune is targeting the NLRP3-ASC pathway using two
complementary approaches, derived from our two technology platforms
(Figure 44):
Figure 44:
Strategy to use our dual proprietary technology platforms to target
NLRP3-ASC
(1) NOD-like receptor protein 3; (2)
Apoptosis-associated speck-like protein containing a CARD, also
PYCARD
Ref: Adapted from Ransohoff
et al., Nature 2017
Small molecule
inhibitors of NLRP3
Leveraging our proprietary
Morphomer platform, the Company has successfully identified and
filed patent applications for various chemical series of potent
small molecule NLRP3 inhibitors. The Company has established
biological activity for these compounds in multiple functional
assays (Figures 45 and 46), and initial animal studies show highly
potent target inhibition in a model of peripheral inflammation
(Figure 47), providing the first evidence of in vivo activity. AC immune is
currently evaluating potential lead compounds for further
in vivo efficacy and
optimization for CNS delivery.
Figure 45:
Screening assay to quantify the compound-mediated inhibition of
IL-1β production in vitro
using human microglia
(1) NOD-like receptor protein 3; (2)
Interleukin 1 beta
Ref: Adapted from Choi et al., Mol Cell 2014
In the figure above, the left
panel illustrates the signal transduction pathway leading to NLRP3
inflammasome activation. The right panel shows the dose dependent
inhibition by a small molecule candidate (cmpd 1) and reference
molecule (Ref) of the NLRP3 pathway post stimulation with
monosodium urate (MSU) crystal that induced interleukin-1β
production by human macrophages. IC50 (inhibition concentration at
50%); DMSO (negative vehicle control) (Ref: AC Immune unpublished
data).
Figure 46:
Secondary assays involving human whole blood demonstrate potent hit
compounds active in vitro
using multiple donors
(1) NOD-like receptor protein 3; (2)
Interleukin 1 beta; (3) Bioluminescence Resonance Energy
Transfer
Ref: AC Immune unpublished
data
Figure 47: In a
mouse model of peritonitis, several of the initial hits targeting
NLRP3 show significant inhibition of IL-1β production in vivo
(1) NOD-like receptor protein 3; (2)
Interleukin 1 beta; (3) Bacterial lipopolysaccaride
Ref: AC Immune unpublished
data
Therapeutic
antibodies for neuroinflammation (mAb-ASC)
It has been shown in the APP/PSI
mouse model of AD, intracellular and extracellular ASC specks are
present. Treatment using an anti-ASC antibody decreases the Abeta
load in these mice (Figure 48).
Figure 48: ASC
specks in AD patients and mouse model of AD
(1) Apoptosis-associated speck-like
protein containing a CARD; (2) Amyloid precursor protein/presenilin
1
Ref:
Vanegas et al., Nature
2017
Using our SupraAntigen platform,
AC Immune generated a novel anti-ASC monoclonal antibodies. It
binds to human and mouse ASC specks with picomolar affinity and
shows specific target engagement on human and mouse macrophages
(arrows in the immunofluorescence images, Figure 49). In addition,
western blot experiments demonstrated target engagement on ASC
specks obtained or purified from recombinant mouse (Rec Mouse) and
recombinant human (Rec Human) while target specificity was
confirmed in human macrophages where ASC is genetically knocked
down (no band for THP-1 ASC KO; Figure 49). Selected antibodies are
currently being evaluated in in
vivo proof-of-concept studies using animal models of human
disease. These have been initiated in Q4 2021 with data estimated
by H2 2022 to validate ASC specks as targets. This would narrow the
antibody candidates for lead declaration. These innovative,
potentially disease-modifying antibodies are designed to have the
highest potential to prevent inflammation and modify the downstream
exacerbation of various proteinopathies.
Figure 49:
Neutralizing anti-ASC antibodies that bind extracellular human ASC
and potently inhibit inflammation-mediated formation of ASC
specks
(1) Apoptosis-associated speck-like
protein containing a CARD; (2) monoclonal antibodies
Ref: AC Immune unpublished
data
TDP-43 imaging diagnostics
To complement our pipeline of PET
imaging tracers, we also selected TDP-43 as a third target. TDP-43
in its physiological function is a protein regulating mRNA
splicing, stability and translation as well as gene transcription.
Similar to Tau, Abeta and a-syn, TDP-43 misfolds in TDP-43
proteinopathies into insoluble aggregates predominantly in the
cytoplasm of neurons, leading to cellular dysfunction and
eventually clinical symptoms. TDP-43 pathology often appears in
other neurodegenerative diseases (e.g., AD) as a part of mixed
pathologies, and it has been proposed that misfolded TDP-43
contributes to the observed clinical phenotype in addition to the
primary pathology. The precise molecular diagnosis and
differentiation of early stages of such diseases is of critical
importance. Using proprietary assays, a set of small molecular
weight compounds from four chemically distinct series were
identified, which bind to patient-derived pathological TDP-43.
Several of these compounds demonstrated favorable pharmacokinetic
profiles in rodents suggesting suitable properties for further
development as PET ligands. We identified candidates showing
nanomolar affinities on TDP-43 aggregates enriched from patients
with TDP-43 proteinopathies. Selected compounds show target
engagement on FTLD-TDP brain sections by high resolution
autoradiography. Affinity and selectivity are being further
optimized to deliver a potential first-in-class PET tracer for
TDP-43.
Figure 50:
First-in-class TDP-43 PET imaging tracer – Discovery Phase
Ref: AC Immune, AD/PD
Conference, 2022
There are no imaging products in
the market today targeting TDP-43. This provides us with a unique
opportunity to become the first company to provide a TDP-43-PET
tracer to the market. We believe the ability to image TDP-43
deposits in the brain will enable fundamental change in the
approach toward treating primary and secondary TDP-43
proteinopathies including improved design for AD clinical trials to
provide the best outcome for patients.
License agreements and
collaborations
Our SupraAntigen and Morphomer
platforms have generated large numbers of clinical assets that
address multiple diseases related to protein misfolding. Selected
key assets in the product pipeline have been licensed for upfront
payments, milestones and royalties to help offset the cost of our
research and internal product development. Discussions with other
companies are ongoing. We have signed a number of licensing
agreements with leading pharmaceutical companies to assist and
accelerate the development of our product pipeline,
including:
|
• |
a worldwide licensing agreement with Genentech signed in
November 2006 (and amended in March 2009, January 2013, May 2014
and May 2015) for crenezumab for AD, under which we may become
eligible to receive payments potentially greater than USD 340 (CHF
314) million, excluding royalties;
|
|
• |
a worldwide licensing agreement with Genentech signed in June
2012 (and amended in December 2015) for anti-Tau antibodies to
treat AD and potentially other indications, under which we may
become eligible to receive payments potentially greater than CHF
400 million, excluding royalties;
|
|
• |
a worldwide licensing agreement with Janssen signed in
December 2014 (and amended in April 2016, July 2017, January 2019
and November 2019) for therapeutic anti-Tau vaccines for AD, and
potentially other Tauopathies, under which we may become eligible
to receive payments totaling up to CHF 500 million, excluding
royalties;
|
|
• |
a worldwide licensing and collaboration agreement (LCA) with
LMI (formerly Piramal Imaging SA) signed in May 2014 for
small-molecule Tau ligands for use as PET tracers under which we
may become eligible to receive payments totaling up to EUR 160 (CHF
167) million, excluding royalties; and
|
|
• |
a worldwide license agreement with Lilly to research and
develop Morphomer Tau small molecules for the treatment of AD and
other neurodegenerative diseases, which was entered into in
December 2018 (and amended in September 2019 and March 2020). The
agreement was deemed effective on January 23, 2019. Under this, we
may become eligible to receive payments up to approximately CHF 1.9
billion, excluding royalties.
|
Further information concerning
details of our agreements and collaborations can be found under
“Item 5: Operating and financial review and prospects.”
Competition
The pharmaceutical and
biopharmaceutical industries are highly competitive across all
therapeutic fields. In the field of neurodegenerative diseases,
there are many public and private companies or institutions that
are actively engaged in the discovery and development of
therapeutic and diagnostic products. Some of these products may
have a similar target to our product candidates or address similar
markets. The industry is still in its infancy in terms of defining
the pathology of neurodegenerative diseases. As disease
understanding progresses, the number of novel product candidates
may well increase and broaden the therapeutic and diagnostic
options in our product markets.
Currently, there is only one
approved disease-modifying product for AD. Most currently approved
therapies seek to treat the symptoms of AD, such as cognitive
decline, but do not slow or stop the progression of the disease. In
addition, commonly, there is off-label prescription of
antidepressant and antipsychotic agents for more patients with
advanced AD who may have agitation, aggressive behaviors, psychosis
and depression.
We expect there to be several
classes of disease-modifying agents that will enter the AD market.
Among our monoclonal antibodies, there are semorinemab targeting
extracellular Tau and crenezumab targeting Abeta oligomers.
Therapeutic vaccines are a second class of disease-modifying
therapies, and include our candidate products ACI-35.030, which
targets aggregated, phosphorylated Tau protein and ACI-24, which
targets oligomeric Abeta. A third class of disease-modifying
therapies, small molecules, include our Morphomer Tau program,
which inhibits Tau aggregates.
The availability of novel
diagnostic agents to visualize the disease development in patients
with AD is critical for successful clinical development of
disease-modifying products in AD. At the forefront of this new
diagnostic effort are PET agents for imaging of disease pathology,
and in particular, Tau-targeting PET agents, which we believe will
allow precise assessment of disease in patients with AD. A similar
situation is developing in other NDD, such as PD, where PET imaging
is becoming available.
ACI-35.030.
ACI-35.030, if approved, would compete with other approved
Tau-targeting therapeutic vaccines. This includes the AADvac1
vaccine developed by Axon Neuroscience, which completed a Phase 2
study in 2019.
ACI-24 for AD.
ACI-24, if approved, would compete with other approved
anti-Abeta-targeting therapeutic vaccines. This includes the ABvac
40 (Araclon Biotech), which is currently being evaluated in a Phase
2 study and UB-311(Vaxxinity), which has completed a Phase 2a
study. In addition, ABvac 42 (Araclon Biotech) has completed a
Phase 1 study and ALZ-101 (Alzinova) is currently being evaluated
in a Phase 1b study.
ACI-24 for DS.
ACI-24 is the first disease-modifying vaccine candidate addressing
DS-related AD, with a potential preventive and therapeutic
application. Although there are symptomatic treatments of DS in
clinical development, to our knowledge there are currently no other
disease-modifying treatments in clinical development for AD in
DS.
ACI-7104. ACI-7104,
if approved, would compete with other approved a-syn-targeting
therapeutic vaccines. This includes the UB-312 vaccine developed by
Vaxxinity, which is being evaluated in a Phase 1 study.
Semorinemab.
Semorinemab is one of several Tau-targeting monoclonal antibodies
in development to potentially act as disease-modifying agents.
Bepranemab (UCB/Roche) and JNJ-63733657 (Janssen) are being
evaluated in Phase 2 studies. BIIB076 (Biogen/Neuroimmune), Lu
AF87908 (Lundbeck), PNT001 (Pinteon Therapeutics), E-2814 (Eisai)
and PRX005 (Prothena/BMS) are being evaluated in Phase 1
studies.
Crenezumab.
Crenezumab is the first monoclonal antibody candidate that targets
Abeta in cognitively healthy individuals at risk of developing
familial AD. However, Lilly’s solanezumab, Roche’s gantenerumab,
lecanemab (BioArctic/Eisai) and donanemab (Eli Lilly) are being
evaluated in studies of presymptomatic AD. ADUHELM (Biogen) has
been approved by FDA under the accelerated approval pathway for the
treatment of patients with mild cognitive impairment and mild
dementia.
Morphomer Tau. AC
Immune has developed the first small molecule targeting aggregated
Tau with high selectivity for the target. In collaboration with
Lilly, this molecule (ACI-3024) was studied in a Phase 1 clinical
trial which was completed in 2020 as a first-in-class,
Tau-specific, disease-modifying, Tau aggregation inhibitor small
molecule for the treatment of neurodegenerative diseases
characterized by misfolded Tau. Together with our partner Eli
Lilly, we have identified optimized candidates that will now be
prioritized for Alzheimer’s disease development. The optimized
candidates have been shown to have enhanced brain uptake, good
safety profiles and high affinity for Tau. These new candidates are
being further characterized in in
vivo preclinical models and will be advanced into
IND-enabling studies, and one of them is expected to advance into
development in 2022. ACI-3024 will continue to be investigated in
Orphan indications.
Tau-PET tracer.
Tauvid (previously known as Flortaucipir) was developed by Eli
Lilly and approved by FDA in May 2020. However, should PI-2620 be
approved, it would also compete with (i) APN-1607 (previously known
as 18F-PM-PBB3), a product candidate in a Phase 2 study and being
advanced by Aprinoia; (ii) 18F-MK-6240, which is being evaluated by
Cerveau/Merck in a Phase 2 clinical trial in patients with ADAD;
(iii) 18F-GTP1, which is being developed by Genentech and is in a
Phase 2 study in subjects at risk of developing ADAD, (iv)
18F-RO6958948, for which Roche has completed a Phase 1 study in
patients with AD and (v)18F-JNJ-067, for which Janssen has
completed a Phase 1 study in patients with AD.
A-syn-PET tracer.
To our knowledge, there are no a-syn PET tracers in the
clinic
A-syn. Several
a-syn antibodies are currently in development; Roche/Prothena
entered a Phase 2 with prasinezumab in June 2017 and in May 2021,
began a Phase 2b study in PD patients with more advanced symptoms;
; Astra Zeneca/Takeda started a Phase 1 study in patients with
Parkinson’s disease with MEDI1341 in August 2020; Lundbeck/Genmab
entered a Phase 2 in Multiple System Atrophy (MSA) with Lu AF82422
in November 2021; AbbVie/BioArctic is preparing for a Phase 2 study
with ABBV-0805; and UCB7853 (UCB/Novartis) entered a Phase 1 study
in December 2020.
TDP-43 antibodies.
To our knowledge, there are no TDP-43 antibodies in the
clinic.
Many of our competitors have
significantly greater financial, technical and human resources than
we have available. Mergers and acquisitions in the pharmaceutical
and biotechnology industries may result in even more resources
being concentrated among a smaller number of our competitors. Our
commercial opportunities and our success will be based in part on
our ability to identify, develop and manage a portfolio of product
candidates that are safer and more effective than competing
products. However, this opportunity could be eroded or even
eliminated if our competitors develop and/or market products that
are novel and have superior safety and efficacy profiles, that may
be brought to the market more rapidly due to greater available
resources, or that are less costly than our current or future
product candidates.
Commercialization strategy
Our strategy to date has been to
focus on identifying partnerships for our early-stage product
candidates as both a way to secure non-dilutive capital to fund our
other research and development programs and also as a way to
accelerate the development of these partnered products by
leveraging our partners’ extensive knowledge in clinical studies,
drug development, manufacturing and commercialization.
With greater financial resources at
our disposal and the significant knowledge acquired by our
scientists and scientific leadership, we intend to retain selected
promising product candidates in-house for a longer period of time
and fund their development from our own resources. This will allow
us to generate greater value from these product candidates,
allowing us to demand more significant terms from a prospective
partner. For example, while we plan to seek a strategic partner for
our Abeta vaccine program in AD, our current plan is to retain full
control of this asset for development in the DS population. We will
commence an upcoming Phase 2 study for AD before potentially
partnering this program and intend to fund further clinical
development in DS from our own financial resources. In the field of
diagnostics, the parallel development of therapeutic compounds and
companion diagnostics is of growing importance to the
pharmaceutical and biopharmaceutical industries. The development
timeframe of a PET diagnostic agent is significantly shorter than
for a therapeutic product, providing the prospect for potential
diagnostic product revenues to be realized quicker than potential
therapeutic product revenues. Our Morphomer platform is
particularly well suited to generate molecules for use in the
development of companion diagnostics.
Given our current stage of product
development, we currently do not have a commercialization
infrastructure. If any of our product candidates is granted
marketing approval, we intend to focus our initial commercial
efforts in the US and select European markets, which we believe
represent the largest market opportunities for us. In those
markets, we expect our commercial operations to include our own
specialty sales force that will target Neurologists and
Gerontologists, both in hospitals and in private practice. In other
markets, we expect to seek partnerships that would maximize our
products’ commercial potential.
Intellectual
property
We strive to protect the
proprietary technology that we believe is important to our
business, including seeking and maintaining US and foreign patents
intended to cover our products and compositions, their methods of
use and processes for their manufacture, and our proprietary
technology platforms, diagnostic candidates and any other
inventions that are commercially important to the development of
our business. We also rely on trade secrets and know-how to protect
aspects of our business that are not amenable to, or that we do not
consider appropriate for, patent protection.
Our success will significantly
depend on our and our collaboration and licensing partners’ ability
to obtain and maintain patent and other proprietary protection for
commercially important technology, inventions and know-how related
to our business, to defend and enforce patents, to preserve the
confidentiality of our trade secrets and to operate our business
without infringing any patents and other intellectual property or
proprietary rights of third parties. See the section titled “Risk
factors— Risks related to intellectual property” for additional
information.
As of December 31, 2021, we owned
or co-owned with our collaboration and licensing partners,
approximately 44 issued US patents and 363 issued patents in other
jurisdictions, as well as 27 pending US patent applications and 519
pending foreign patent applications. As of December 31, 2021, we
licensed approximately 28 issued US patents and 257 issued patents
in other jurisdictions, as well as 19 pending US patent
applications and 333 pending foreign patent applications.
The patent portfolios for our most
advanced product candidates as of December 31, 2021 are summarized
below:
Anti-Tau vaccines
Our patent portfolio for anti-Tau
vaccines includes a patent family with composition-of-matter claims
(including claims directed to the ACI-35 antigenic peptide and a
pharmaceutical composition comprising such an antigenic peptide),
claims directed to treating certain indications using ACI-35
including AD, and claims directed to using ACI-35 to induce an
immune response. This patent family currently contains
approximately 28 issued patents and two pending patent applications
in 27 countries. The issued patents in this patent family, if the
appropriate maintenance, renewal, annuity or other governmental
fees are paid, are expected to expire in 2030, excluding any
additional term for patent term adjustments or patent term
extensions.
Our patent portfolio for anti-Tau
vaccines also includes a patent family relating to therapeutic Tau
vaccine claims (including claims directed to a pharmaceutical
composition comprising an antigenic Tau peptide), claims directed
to using such vaccines to induce an immune response in a subject,
and claims directed to methods for preventing or treating a
neurodegenerative disease or disorder, including AD, among others.
This patent family currently contains 1 issued patent and
approximately forty pending patent applications in 38
countries. The issued patent and any patents issuing in this
patent family, if the appropriate maintenance, renewal, annuity or
other governmental fees are paid, are expected to expire in 2038,
excluding any additional term for patent term adjustments or patent
term extensions.
ACI-24
Our patent portfolio for ACI-24
includes a patent family with composition-of-matter claims
(including claims directed to the ACI-24 antigenic construct),
claims directed to treating certain indications using ACI-24
including AD, and claims directed to using ACI-24 to induce an
immune response. This patent family currently contains
approximately 26 issued patents and 8 pending patent applications
in 30 countries. With respect to the US, we own two issued US
patents. The issued patents in this patent family, if the
appropriate maintenance, renewal, annuity or other governmental
fees are paid, are expected to expire in 2026, excluding any
additional term for patent term adjustments or patent term
extensions.
Our patent portfolio for ACI-24
also includes a patent family directed to the use of the ACI-24
vaccine in the treatment and/or prevention of memory and/or
cognitive impairments or abnormalities in the DS population, among
others. As of December 31, 2021, in this patent family, we owned
approximately 12 issued patents and 6 pending patent applications
in 18 countries. Issued patents in this patent family, if the
appropriate maintenance, renewal, annuity or other governmental
fees are paid, are expected to expire in 2032, excluding any
additional term for patent term adjustments or patent term
extensions.
Our patent portfolio for ACI-24
also includes a patent family relating to therapeutic anti-Abeta
vaccine claims (including claims directed to a pharmaceutical
composition comprising an antigenic peptide), and claims directed
to using such vaccines in treating, preventing, inducing a
protective immune response against or alleviating the symptoms
associated with an Abeta-associated disease in a subject, among
others. This patent family currently contains one issued US patent
and approximately 33 pending patent applications in 32 countries.
Any issued patents in this patent family, if the appropriate
maintenance, renewal, annuity or other governmental fees are paid,
are expected to expire in 2039, excluding any additional term for
patent term adjustments or patent term extensions.
ACI-7104
Our patent portfolio relating to
ACI-7104 includes patents and patent applications with
composition-of-matter claims (including claims directed to the
peptide, as well as pharmaceutical formulations comprising the
peptide), and claims directed to the use of compounds comprising
the peptide in treating or preventing synucleinopathies including
PD and MSA.
Our patent portfolio relating to
ACI-7104 includes patents and patent applications that we own in
two different patent families. As of December 31, 2021, in these
patent families, we owned approximately 14 issued patents and
15 pending patent applications, in 11 countries. With respect to
the US, we owned two issued US patents. Issued patents in the basic
patent family, if the appropriate maintenance, renewal, annuity or
other governmental fees are paid, are expected to expire in 2029,
excluding any additional term for patent term adjustments or patent
term extensions.
Semorinemab
Our global patent portfolio
relating to semorinemab includes patents and patent applications
with claims directed to compositions of matter, methods of
treatment for certain indications including AD, and methods of use,
among others.
Crenezumab
Our patent portfolio relating to
crenezumab includes patents and patent applications with claims
directed to composition of matter (including claims directed to the
crenezumab antibody or a fragment thereof, a polynucleotide
encoding the crenezumab antibody or a fragment thereof, a cell line
used to produce the crenezumab antibody as well as pharmaceutical
compositions comprising the crenezumab antibody), claims directed
to treating certain indications using the crenezumab antibody
including AD, claims directed to a method of manufacturing the
crenezumab antibody and claims directed to diagnostic and
prognostic uses of the crenezumab antibody.
Our patent portfolio relating to
crenezumab includes patents and patent applications that we own or
co-own in four different patent families. As of December 31, 2021,
we owned or co-owned approximately 49 patents (not including the
patents in the individual countries where the issued European
patent was validated) and 15 patent applications in 34 countries in
our main patent family directed to the crenezumab antibody and
methods of using the crenezumab antibody to treat certain
indications, including AD. This patent portfolio includes three
issued US patents and one pending US patent applications, which, if
the appropriate maintenance or other governmental fees are paid,
are expected to expire in 2027, excluding any additional term for
patent term adjustments or patent term extensions. This patent
portfolio also includes a PCT patent application that was filed on
July 13, 2007. If the appropriate maintenance, renewal, annuity, or
other governmental fees are paid, national-stage applications
issuing from this PCT patent application are expected to expire in
2027, excluding any additional term for patent term adjustments or
patent term extensions, as applicable.
Morphomer Tau
Our patent portfolio relating to
Morphomer Tau therapeutics includes patent applications with claims
directed to composition of matter (including claims directed to the
molecule, a pharmaceutical composition comprising such molecule and
a mixture comprising such molecule), and claims directed to
prevention and treatment of certain indications using such
molecules including AD and PSP, among others.
Our patent portfolio relating to
the Morphomer Tau therapeutic program includes patent applications
that we own or co-own in four different patent families. As of
December 31, 2021, we owned or co-owned approximately 49 pending
patent applications and one US issued patent in our main patent
family directed to the ACI-3024 small molecule Tau aggregation
inhibitor. If the appropriate maintenance, renewal, annuity, or
other governmental fees are paid, national-stage applications
issuing from this PCT patent application are expected to expire in
2039, excluding any additional term for patent term adjustments or
patent term extensions, as applicable.
PI-2620
Our patent portfolio relating to
PI-2620 includes patent applications with claims directed to
composition of matter (including claims directed to the molecule,
its precursor and a diagnostic composition comprising such
molecule), claims directed to diagnosis of certain indications
using PI-2620 including AD and PSP, and claims directed to a method
of manufacturing PI-2620, among others.
Our patent portfolio relating to
PI-2620 includes patent applications that we own or co-own in three
different patent families. As of December 31, 2021, we owned or
co-owned 2 patents and approximately 14 patent applications in 16
countries in our main patent family directed to the PI-2620
molecule, its precursor and methods of using the PI-2620 to
diagnose certain indications, including AD and PSP. This main
patent family includes one issued US patent If the appropriate
maintenance, renewal, annuity, or other governmental fees are paid,
national-stage applications issuing from this PCT patent
application are expected to expire in 2037, excluding any
additional term for patent term adjustments or patent term
extensions, as applicable.
A-syn
PET Tracer
Our patent portfolio relating to
a-syn diagnostics includes composition of matter claims (including
claims directed to the ACI-12589 molecule, its precursor, and
diagnostic compositions comprising the molecule), and claims
directed to use of the molecule in imaging and in diagnostics of
a-synucleinopathies including PD and MSA.
Our patent portfolio relating to
a-syn diagnostics includes patents and patent applications that we
own in three different patent families. If the appropriate
maintenance, renewal, annuity or other governmental fees are paid,
any issued patents are expected to provide protection up to 2041,
excluding any additional term for patent term adjustments or patent
term extensions, as applicable.
Manufacturing
and supply
We do not own or operate facilities
for the manufacture, packaging, labeling, storage, testing or
distribution of preclinical or clinical supplies of any of our
product candidates. We instead contract with and rely on
third-party CMOs to manufacture, package, label, store, test and
distribute all preclinical development and clinical supplies of our
product candidates, and we plan to continue to do so for the
foreseeable future. We have established relationships with CMOs
such as WuXi AppTec (WuXi STA), WuXi Biologics, Avecia, Almac
Clinical Services, Bachem AG, Evonik Industries AG, Polymun
Scientific Immunbiologische Forschung GmbH, piCHEM Forschungs-und
Entwicklungs GmbH, Baccinex SA, Solvias AG and Pfenex Inc. among
others.
Compliance with governing rules and quality requirements
The facilities used by our
collaboration partners and CMOs to manufacture our product
candidates are systematically audited by local authorities and
occasionally inspected by competent authorities where the clinical
studies are ongoing. The facilities where the commercial
productions are performed will have to be approved by the FDA or
other relevant regulatory authorities, pursuant to inspections that
are conducted after we submit our NDA or comparable marketing
applications. We perform periodic quality audits of the
manufacturing facilities and CMOs to monitor their compliance with
the regional laws, regulations and applicable cGMP standards and
other laws and regulations, such as those related to environmental
health and safety matters. The scope of our audits also involves
monitoring the ability of our providers to maintain adequate QCs
and QA systems including personnel qualification.
After manufacturing, our products
are submitted to extensive characterization and QC testing plans
performed by using properly developed analytical methods that are
qualified or validated; this ensures the accuracy of the results
generated and provides evidence of the quality of our products. In
addition, our products are submitted to detailed and standardized
stability programs aimed at demonstrating product stability during
the storage period; this, in addition to guaranteeing the safety of
the products, supports the definition of a suitable supply chain
that may encompass the distribution of the products in different
continents.
Contractual framework
We have established, with CMOs
supplying active pharmaceutical ingredients, drug substances or
drug products under cGMP, quality agreements and master service
agreements. Quality agreements define the quality standards
required to develop, produce and supply the product, and also
define the responsibilities related to the collaboration with
regards to the quality related aspects. Manufacturing service
agreements define the commercial and financial framework under
which product manufacturing under cGMP is performed. Any failure to
achieve and maintain compliance with the laws, regulations and
standards, suspension of the manufacturing of our product
candidates or revoke of cGMP permissions, which would adversely
affect our business and reputation, are defined in the master
service agreements and quality agreements. The risk that any
third-party providers may breach the agreements they have with us
because of factors beyond our control and the possibility that they
may also terminate or refuse to renew their agreements because of
their own financial difficulties or business priorities,
potentially at a time that is costly or otherwise inconvenient for
us, is managed by us with constant investments toward maintaining
reserve stocks and in-depth process know-how. The latter is
supported by continuous in-house process development and production
activities of small-scale/research grade materials, which may offer
the chance to rapidly identify alternative contract manufacturers
to whom the manufacturing process could be transferred providing
continuity for the clinical study.
Interaction with CMOs
Finally, our partnerships with CMOs
are managed through an efficient project management platform in
which teams are formed with the representatives of each key
function from both parties. Meetings occur either through telephone
conferences aimed at updating short-term actions or face-to-face
conferences when mid- to long-term development plans are
discussed.
Government
regulation and our regulatory department
Our regulatory department has a
strong culture of regulatory compliance, operating under three
guiding principles, to:
|
• |
provide constructive regulatory input for development
products;
|
|
• |
ensure smooth regulatory approvals by anticipating hurdles;
and
|
|
• |
build confidence with regulators by continuous
communication.
|
The QA group is included within the regulatory department with
the mission to:
|
• |
create and maintain a corporate quality management system;
and
|
|
• |
ensure cGCP, cGMP, cGLP and current Good Distribution Practice
(cGDP) compliance.
|
A science-driven approach is the
cornerstone of our interactions and this has helped us to build and
maintain a high level of trust with regulators. Besides informal
conversations with the authorities, our regulatory department has
conducted several pre-Investigational New Drug (pre-IND), Type B
and Type C meetings with the FDA (ACI-24 for AD and DS, ACI-7104
and PI-2620) and Scientific Advice meetings, which are the European
equivalent of pre-IND meetings (with the German
Paul-Ehrlich-Institut, Swedish Medical Products Agency; UK Medicine
& Healthcare Products Regulatory Agency, Finnish Medicines
Agency, the Spanish Agency of Medicines and Medical Devices and the
EMA). Since 2008, our regulatory department has filed a total of 19
clinical trial applications in the EU (one each in Austria,
Denmark, the Netherlands and Poland, two in Germany, three in
Sweden, four in Finland and five in the UK) and 4 INDs in the US.
Given the seriousness of AD and public pressure for new
therapeutics, we consider regulatory agencies to be important
stakeholders in our product development strategies. We are
committed to working closely with global regulatory authorities to
adhere to and achieve the highest levels of safety and quality of
our product candidates in the most timely and efficient manner. The
transparency we have achieved and our goal of a close working
relationship with the regulatory agencies, in particular the FDA
and the EMA, are intended to facilitate expeditious execution
through the regulatory approval process.
Our regulatory department contains
a QA group. As every quality issue ultimately requires regulatory
involvement and input, this approach is intended to lead to rapid
resolution of issues and ensure full compliance to satisfy both the
reviewers and the inspectors at the government health authorities.
Our regulatory department is charged with keeping our entire
organization, directly or indirectly involved in the clinical study
application process, in a state of “inspection readiness.” To that
end, we ensure that the Trial Master Files are complete and
regularly updated. Our regulatory department is also tasked with
generating our annual quality plan. The personnel tasked with QA
have issued a set of approximately 90 standard operating procedures
and working instructions and continuously train the relevant staff.
Our QA personnel conduct regular audits, including in-person audits
of the contract manufacturers, contract research organizations and
laboratories conducting primary endpoint analysis. In addition, we
have a full-time QA documentation assistant to ensure good
documentation practice and archiving.
Product
approval process
The clinical studies,
manufacturing, labeling, storage, distribution, record-keeping,
advertising, promotion, import, export and marketing, among other
things, of our product candidates are subject to extensive
regulation by governmental authorities in the US and other
countries. The US FDA, under the Federal Food, Drug, and Cosmetic
Act (FDCA), regulates pharmaceutical products in the US. The steps
required before a drug may be approved for marketing in the US
generally include:
|
• |
the completion of preclinical laboratory tests and animal
tests conducted under cGLP regulations;
|
|
• |
the submission to the FDA of an IND application for human
clinical testing, which must become effective before human clinical
studies commence;
|
|
• |
obtaining a positive opinion from the ethics committee
(Europe)/institutional review board (US) to commence study on human
subjects;
|
|
• |
the performance of adequate and well-controlled human clinical
studies to establish the safety and efficacy of the product
candidate for each proposed indication and conducted in accordance
with cGCP requirements;
|
|
• |
pre-NDA submission meeting with FDA (highly
recommended);
|
|
• |
the submission to the FDA of an NDA;
|
|
• |
the FDA’s acceptance of the NDA;
|
|
• |
satisfactory completion of an FDA Pre-Approval Inspection
(PAI) of the manufacturing facilities at which the product is made
to assess compliance with cGMP requirements;
|
|
• |
the FDA’s review and approval of an NDA prior to any
commercial marketing or sale of the drug in the US; and
|
|
• |
having parallel scientific advice from the EMA or Health
Technology Assessment body whereby the payors are involved at the
outset (Phase 2), which is intended to facilitate the design of
clinical studies to target primarily populations with a high chance
of obtaining reimbursement and accelerate the process of time to
reimbursement.
|
The FDA has various programs,
including Fast Track, Priority review, Accelerated Approval and
Breakthrough Therapy designation, which are intended to increase
agency interactions, expedite or facilitate the process for
reviewing product candidates, and/or provide for initial approval
based on surrogate endpoints. We believe that one or more of our
product candidates may qualify for some of these expedited
development and review programs. However, even if a product
candidate qualifies for one or more of these programs, the FDA may
later decide that the product candidate no longer meets the
conditions for qualification.
The Fast Track program is intended
to expedite or facilitate the process for reviewing new drugs that
meet certain criteria. Specifically, new drugs are eligible for
Fast Track designation if they are designed to treat a serious or
life-threatening condition and demonstrate the potential to address
unmet medical needs for the condition. Fast Track designation
applies to the combination of the product and the specific
indication for which it is being studied. The sponsor of a new drug
may request the FDA to designate the drug as a Fast Track product
at any time during the clinical development of the product. AD, for
example, meets both pre-requisites—it is life-threatening and
constitutes an unmet medical need. Unique to a Fast Track product,
the FDA may consider for review sections of the marketing
application on a rolling basis before the complete application is
submitted if the sponsor provides a schedule for the submission of
the sections of the application, the FDA agrees to accept sections
of the application and determines that the schedule is acceptable,
and the sponsor pays any required user fees upon submission of the
first section of the application.
Any product submitted to the FDA
for marketing, including under a Fast Track program may be eligible
for other types of FDA programs intended to expedite development
and review, such as Priority Review and Accelerated Approval. Any
product is eligible for priority review if it has the potential to
provide safe and effective therapy where no satisfactory
alternative therapy exists or it provides a significant improvement
in the treatment, diagnosis or prevention of a disease compared
with marketed products. The FDA will attempt to direct additional
resources to the evaluation of an application for a new drug
designated for Priority Review to facilitate the review.
Additionally, a product may be eligible for the Accelerated
Approval program. Product candidates that are studied for their
safety and effectiveness in treating serious or life-threatening
illnesses and that provide meaningful therapeutic benefit over
existing treatments may receive Accelerated Approval, which means
that they may be approved on the basis of adequate and
well-controlled clinical studies establishing that the product has
an effect on a surrogate endpoint that is reasonably likely to
predict a clinical benefit, or on the basis of an effect on a
clinical endpoint other than survival or irreversible morbidity. As
a condition of approval, the FDA may require that a sponsor of a
drug receiving Accelerated Approval perform adequate and
well-controlled post-marketing clinical studies. Failure to conduct
required post-approval trials, or the inability to confirm a
clinical benefit during post-marketing trials, may allow the FDA to
withdraw the drug from the market on an expedited basis. In
addition, as a condition for Accelerated Approval the FDA currently
requires pre-approval of promotional materials, which could
adversely impact the timing of the commercial launch of the
product. The Fast Track, Priority Review and Accelerated Approval
programs do not change the standards for approval but may expedite
the development or approval process.
The Food and Drug Administration
Safety and Innovation Act of 2012 also amended the FDCA to require
the FDA to expedite the development and review of a breakthrough
therapy. A drug can be designated as a breakthrough therapy if it
is intended to treat a serious or life-threatening disease or
condition and preliminary clinical evidence indicates that it may
demonstrate substantial improvement over existing therapies in one
or more clinically significant endpoints. A sponsor may request
that a drug be designated as a breakthrough therapy at any time
during the clinical development of the product. If so designated,
the FDA shall act to expedite the development and review of the
product’s marketing application, including by meeting with the
sponsor throughout the product’s development, providing timely
advice to the sponsor to ensure that the development program to
gather nonclinical and clinical data is as efficient as
practicable, involving senior managers and experienced review staff
in a cross-disciplinary review, assigning a cross-disciplinary
project lead for the FDA review team to facilitate an efficient
review of the development program and to serve as a scientific
liaison between the review team and the sponsor, and taking steps
to ensure that the design of the clinical trials is as efficient as
practicable.
The testing and approval process
requires substantial time, effort and financial resources, and the
receipt and timing of any approval is uncertain. Given this
paradigm, AD has been given Life-Threatening Disease status by the
FDA and therefore AD therapies are eligible for the expanded access
program for investigational drugs and other pathways such as
Breakthrough Therapy, Accelerated Approval and Priority Review.
Additionally, a single well-designed, well-conducted, pivotal
clinical study could be sufficient to trigger market approval
pending a successful PAI.
Preclinical studies include
laboratory evaluations of the product candidate, as well as animal
studies to assess the potential safety and efficacy of the product
candidate. The results of the preclinical studies, together with
manufacturing information and analytical data, are submitted to the
FDA as part of the IND, which must become effective before clinical
studies may be commenced. The IND will automatically become
effective 30 days after receipt by the FDA, unless the FDA raises
concerns or questions about the conduct of the studies as outlined
in the IND prior to that time. In this case, the IND sponsor and
the FDA must resolve any outstanding concerns before clinical
studies can proceed.
Clinical studies involve the
administration of the product candidates to healthy volunteers or
patients with the disease to be treated under the supervision of a
qualified principal investigator. Clinical studies are conducted
under protocols detailing, among other things, the objectives of
the study, the parameters to be used in monitoring safety and the
efficacy criteria to be evaluated. A protocol for each clinical
study and any subsequent protocol amendments must be submitted to
the FDA as part of the IND. Further, each clinical study must be
reviewed and approved by an independent IRB, either centrally or
individually at each institution at which the clinical study will
be conducted. The IRB will consider, among other things, ethical
factors, the safety of human subjects and the possible liability of
the institution. There are also requirements governing the
reporting of ongoing clinical studies and clinical study results to
public registries. The FDA, the IRB or the clinical study sponsor
may suspend or terminate clinical studies at any time on various
grounds, including a finding that the subjects or patients are
being exposed to an unacceptable health risk. Additionally, some
clinical studies are overseen by an independent group of qualified
experts organized by the clinical study sponsor, known as a Data
Safety Monitoring Board/Committee. This group provides
authorization for whether or not a study may move forward at
designated checkpoints based on access to certain data from the
study. We may also suspend or terminate a clinical study based on
evolving business objectives and/or competitive climate.
Clinical studies are typically
conducted in three sequential phases prior to approval, but the
phases may overlap. These phases generally include the
following:
Phase 1. Phase 1 clinical studies
represent the initial introduction of a product candidate into
human subjects, frequently healthy volunteers. In Phase 1, the
product candidate is usually tested for safety, including adverse
effects, dosage tolerance, absorption, distribution, metabolism,
excretion and pharmacodynamics.
Phase 2. Phase 2 clinical studies
usually involve studies in a limited patient population to (i)
evaluate the efficacy of the product candidate for specific
indications, (ii) determine dosage tolerance and optimal dosage,
and (iii) identify possible adverse effects and safety risks.
Phase 3. If a product candidate is
found to be potentially effective and to have an acceptable safety
profile in Phase 2 studies, the clinical study program will be
expanded to Phase 3 clinical studies to further demonstrate
clinical efficacy, optimal dosage and safety within an expanded
patient population at geographically dispersed clinical study
sites.
Phase 4. Phase 4 clinical studies
are conducted after approval to gain additional experience from the
treatment of patients in the intended therapeutic indication and to
document a clinical benefit in the case of drugs approved under
Accelerated Approval regulations, or when otherwise requested by
the FDA in the form of post-marketing requirements or commitments.
Failure to conduct any required Phase 4 clinical studies promptly
could result in withdrawal of approval.
The results of preclinical studies
and clinical studies, including negative or ambiguous results as
well as positive findings, together with detailed information on
the manufacture, composition and quality of the product, are
submitted to the FDA in the form of an NDA requesting approval to
market the product. The NDA must be accompanied by a significant
user-fee payment. The FDA has substantial discretion in the
approval process and may refuse to accept any application or decide
that the data is insufficient for approval and require additional
preclinical, clinical or other studies.
We estimate that it generally takes
10 to 15 years, or possibly longer, to discover, develop and bring
to market a new pharmaceutical or biopharmaceutical product in the
US. Several years may be needed to complete each phase, including
discovery, preclinical, Phase 1, 2 or 3, or marketing
authorization.
In addition, under the Pediatric
Research Equity Act, an NDA or supplement to an NDA must contain
data to assess the safety and effectiveness of the drug for the
claimed indications in all relevant pediatric subpopulations and to
support dosing and administration for each pediatric subpopulation
for which the product is safe and effective. Recently, the Food and
Drug Administration Safety and Innovation Act (FDASIA), which was
signed into law on July 9, 2012, amended the FDCA. The FDASIA
requires that a sponsor who is planning to submit a marketing
application for a drug or biological product that includes a new
active ingredient, new indication, new dosage form, new dosing
regimen or new route of administration submit an initial Pediatric
Study Plan within 60 days of an end-of-Phase-2 meeting or as may be
agreed between the sponsor and the FDA. The initial Pediatric Study
Plan must include an outline of the pediatric study or studies that
the sponsor plans to conduct, including study objectives and
design, age groups, relevant endpoints and statistical approach, or
a justification for not including such detailed information, and
any request for a deferral of pediatric assessments or a full or
partial waiver of the requirement to provide data from pediatric
studies along with supporting information. The FDA and the sponsor
must reach agreement on the Pediatric Study Plan. A sponsor can
submit amendments to an agreed-upon initial Pediatric Study Plan at
any time if changes to the pediatric plan need to be considered
based on data collected from nonclinical studies, early-phase
clinical trials, and/or other clinical development programs.
The cost of preparing and
submitting an NDA is substantial. Under federal law, NDAs are
subject to substantial application user fees and the sponsor of an
approved NDA is also subject to annual product and establishment
user fees. Under the Prescription Drug User Fee Act (PDUFA), as
amended, each NDA must be accompanied by a user fee. The FDA
adjusts the PDUFA user fees on an annual basis. PDUFA VI eliminates
fees for supplements as well as for establishments, although
applicants will be assessed for annual prescription drug program
fees for prescription drug products, rather than the prescription
drug product fee assessed under the previous iteration of PDUFA.
According to the FDA’s fee schedule for the 2022 FY, the user fee
for each NDA application requiring clinical data is USD 3,117,218
and the annual program fee is USD 369,413. Fee waivers or
reductions are available in certain circumstances, including a
waiver of the application fee for the first application filed by a
small business. Additionally, no user fees are assessed on NDAs for
products designated as orphan drugs, unless the product also
includes a non-orphan indication.
Once the NDA submission has been
submitted, the FDA has 60 days after submission of the NDA to
conduct an initial review to determine whether it is sufficient to
accept for filing. Under the PDUFA, the FDA sets a goal date by
which it plans to complete its review. This is typically 12 months
from the date of submission of the NDA application. The review
process is often extended by FDA requests for additional
information or clarification. Before approving an NDA, the FDA will
inspect the facilities at which the product is manufactured and
will not approve the product unless the manufacturing facility
complies with cGMP regulations and may also inspect clinical study
sites for integrity of the data supporting safety and efficacy. The
FDA may also convene an advisory committee of external experts to
provide input on certain review issues relating to risk, benefit
and interpretation of clinical study data. The FDA is not bound by
the recommendations of an advisory committee, but generally follows
such recommendations in making its decisions. The FDA may delay
approval of an NDA if applicable regulatory criteria are not
satisfied and/or the FDA requires additional testing or
information. The FDA may require post-marketing testing and
surveillance to monitor safety or efficacy of a product.
After the FDA evaluates the NDA and
conducts inspections of the manufacturing facilities where the drug
product and/or its API will be produced, it may issue an Approval
Letter or a Complete Response Letter. An Approval Letter authorizes
commercial marketing of the drug with specific prescribing
information for specific indications. A Complete Response Letter
indicates that the review cycle of the application is complete and
the application is not ready for approval. A Complete Response
Letter may require additional clinical data and/or an additional
pivotal Phase 3 clinical study or studies, and/or other
significant, expensive and time-consuming requirements related to
clinical studies, preclinical studies or manufacturing. Even if
such additional information is submitted, the FDA may ultimately
decide that the NDA does not satisfy the criteria for approval. The
FDA could also approve the NDA with a Risk Evaluation and
Mitigation Strategy (REMS), plan to mitigate risks, which could
include medication guides, physician communication plans, or
elements to assure safe use, such as restricted distribution
methods, patient registries and other risk minimization tools. The
FDA also may condition approval on, among other things, changes to
proposed labeling, development of adequate controls and
specifications, or a commitment to conduct one or more
post-marketing studies or clinical studies. Such post-marketing
testing may include Phase 4 clinical studies and surveillance to
further assess and monitor the product’s safety and effectiveness
after commercialization.
Special
protocol assessment
The FDA and an IND sponsor may
agree in writing on the design and size of clinical studies
intended to form the primary basis of a claim of effectiveness in
an NDA. This process is known as a special protocol assessment
(SPA). Upon a specific request for a SPA by an IND sponsor, the FDA
will evaluate the protocol. If an SPA agreement is reached,
however, it is not a guarantee of product approval by the FDA or
approval of any permissible claims about the product. The FDA
retains significant latitude and discretion in interpreting the
terms of the SPA agreement and the data and results from any study
that is the subject of the SPA agreement. In particular, the SPA
agreement is not binding on the FDA if previously unrecognized
public health concerns later come to light, other new scientific
concerns regarding product safety or efficacy arise, the IND
sponsor fails to comply with the agreed-upon protocol, or the
relevant data, assumptions, or information provided by the IND
sponsor when requesting a SPA agreement change, are found to be
false statements or misstatements, or are found to omit relevant
facts. An SPA agreement may not be changed by the sponsor or the
FDA after the study begins except with the written agreement of the
sponsor and the FDA, or if the FDA determines that a substantial
scientific issue essential to determining the safety or
effectiveness of the drug was identified after the testing
began.
Orphan-drug designation
Under the Orphan Drug Act, the FDA
may grant orphan designation to a drug or biological product
intended to treat a rare disease or condition, which is a disease
or condition that either affects fewer than 200,000 individuals in
the US, or affects more than 200,000 individuals in the US but
there is no reasonable expectation that the cost of developing and
making a drug product available in the US for this type of disease
or condition will be recovered from sales of the product in the US.
Orphan-product designation must be requested before submitting an
NDA. After the FDA grants orphan-product designation, the identity
of the therapeutic agent and its potential orphan use are disclosed
publicly by the FDA. Orphan-product designation does not convey any
advantage in or shorten the duration of the regulatory review and
approval process.
If a product that has orphan
designation subsequently receives the first FDA approval for the
disease or condition for which it has such designation, the product
is entitled to orphan-product exclusivity, which means that the FDA
cannot approve any other applications to market the same drug or
biological product for the same indication for 7 years, except in
limited circumstances, such as a showing of clinical superiority to
the product with orphan exclusivity. The designation of such drug
also entitles a party to financial incentives such as opportunities
for grant funding toward clinical study costs, tax advantages and
user-fee waivers. Competitors, however, may receive approval of
different products for the indication for which the orphan product
has exclusivity or obtain approval for the same product but for a
different indication for which the orphan product has exclusivity.
Orphan-product exclusivity also could block the approval of one of
our products for 7 years if a competitor obtains approval of the
same drug or biological product as defined by the FDA or if our
product candidate is determined to be contained within the
competitor’s product for the same indication or disease. If a drug
product designated as an orphan product receives marketing approval
for an indication broader than what is designated, it may not be
entitled to orphan-product exclusivity. Orphan-drug status in the
EU has similar but not identical benefits in that
jurisdiction.
Disclosure of clinical trial information
Sponsors of clinical trials (other than Phase 1 trials) of
FDA-regulated products, including drugs, are required to register
and disclose certain clinical trial information. Information
related to the product, comparator, patient population, phase of
investigation, trial sites and investigators and other aspects of
the clinical trial is made public as part of the registration.
Sponsors are also obligated to disclose the results of their
clinical trials after completion. Disclosure of the results of
certain trials may be delayed until the new product or new
indication being studied has been approved. However, there are
evolving rules and increasing requirements for publication of
trial-related information, and it is possible that data and other
information from trials involving drugs that never garner approval
could be required to be disclosed in the future. In addition,
publication policies of major medical journals mandate certain
registration and disclosures as a pre-condition for potential
publication, even when this is not presently mandated as a matter
of law. Competitors may use this publicly available information to
gain knowledge regarding the progress of development
programs.
Post-approval requirements
Drugs manufactured or distributed
pursuant to FDA approvals are subject to pervasive and continuing
regulation by the FDA, including, among other things, requirements
relating to record-keeping, periodic reporting, product
distribution, advertising and promotion and reporting of adverse
experiences with the product. After approval, most changes to the
approved product, such as adding new indications or other labeling
claims, are subject to prior FDA review and approval. There also
are continuing, annual user-fee requirements for any marketed
products and the establishments at which such products are
manufactured, as well as new application fees for supplemental
applications with clinical data.
In addition, drug manufacturers and
other entities involved in the manufacture and distribution of
approved drugs are required to register their establishments with
the FDA and state agencies, and are subject to periodic unannounced
inspections by the FDA and these state agencies for compliance with
cGMP requirements. Changes to the manufacturing process are
strictly regulated and often require prior FDA approval before
being implemented. FDA regulations also require investigation and
correction of any deviations from cGMP and impose reporting and
documentation requirements upon the sponsor and any third-party
manufacturers that the sponsor may decide to use. Accordingly,
manufacturers must continue to expend time, money, and effort in
the areas of production and QC to maintain cGMP compliance.
Once an approval is granted, the
FDA may withdraw the approval if compliance with regulatory
requirements and standards is not maintained or if problems occur
after the product reaches the market. Later discovery of previously
unknown problems with a product, including AEs of unanticipated
severity or frequency, or with manufacturing processes, or failure
to comply with regulatory requirements, may result in revisions to
the approved labeling to add new safety information, imposition of
post-marketing studies or clinical studies to assess new safety
risks, or imposition of distribution or other restrictions under a
REMS program. Other potential consequences include, among other
things:
|
• |
restrictions on the marketing or manufacturing of the product,
complete withdrawal of the product from the market or product
recalls;
|
|
• |
fines, warning letters or holds on post-approval clinical
studies;
|
|
• |
refusal of the FDA to approve pending NDAs or supplements to
approved NDAs, or suspension or revocation of product license
approvals;
|
|
• |
product seizure or detention, or refusal to permit the import
or export of products; or
|
|
• |
injunctions or the imposition of civil or criminal
penalties.
|
The FDA strictly regulates
marketing, labeling, advertising and promotion of products that are
placed on the market. Drugs may be promoted only for the approved
indications and in accordance with the provisions of the approved
label. The FDA and other agencies actively enforce the laws and
regulations prohibiting the promotion of off-label uses, and a
company that is found to have improperly promoted off-label uses
may be subject to significant liability.
Patent
term restoration and marketing exclusivity
Depending upon the timing, duration, and specifics of FDA
approval of the use of our product candidates, some of our US
patents may be eligible for limited patent term extension under the
Drug Price Competition and Patent Term Restoration Act of 1984,
referred to as the Hatch-Waxman Act. The Hatch-Waxman Act permits a
patent term to be extended up to 5 years as compensation for patent
term effectively lost due to the FDA’s pre-market approval
requirements. However, patent term restoration cannot extend the
remaining term of a patent beyond a total of 14 years from the
product’s approval date. The patent term restoration period is
generally one-half of the time between the effective date of an IND
and the submission date of an NDA, plus the time between the
submission date of an NDA and the approval of that application,
except that the review period is reduced by any time during which
the applicant failed to exercise due diligence. Only one patent
applicable to an approved drug is eligible for the extension.
Extensions are not granted as a matter of right and the extension
must be applied for prior to expiration of the patent and within a
60-day period from the date the product is first approved for
commercial marketing. The USPTO, in consultation with the FDA,
reviews and approves the application for any patent term extension
or restoration. Where a product contains multiple active
ingredients, if any one active ingredient has not been previously
approved, it can form the basis of an extension of patent term
provided the patent claims that ingredient or the combination
containing it.
In the future, we may apply for
patent term restoration for some of our presently owned patents to
add patent life beyond their current expiration date, depending on
the expected length of clinical studies and other factors involved
in the submission of the relevant NDA; however, there can be no
assurance that any such extension will be granted to us.
The Biologics Price Competition and
Innovation Act of 2009 provides up to 12 years of non-patent data
exclusivity within the US to the first applicant to gain approval
of a Biologics License Application for a new biologic product that
has not previously been approved by the FDA, which we refer to as a
reference product. This 12-year data exclusivity does prohibit the
FDA from approving a biosimilar or interchangeable product of such
reference product until 12 years after the licensure of such
reference product. In addition, the FDA will not accept a
biosimilar or interchangeable product application for review until
4 years after the date of first licensure of such reference
product. Under 21CFR314.108, 5 years’ exclusivity is also granted
to new chemical entities that contain no active moiety that has
been approved by the FDA under section 505(b). This market
exclusivity bars the FDA from accepting for review any ANDA or
505(b)(2) application for a drug containing the same active moiety
for (i) 5 years if an ANDA or 505(b)(2) application does not
contain a paragraph IV certification to a listed patent, or (ii) 4
years if an ANDA or 505(b)(2) is submitted containing a paragraph
IV certification to a listed patent. Moreover, pediatric
exclusivity, if granted, may add 6 months of exclusivity if the
reference product has been studied with respect to a pediatric
indication in accordance with certain regulatory requirements. A
reference product may also be granted 7 years of orphan-drug
exclusivity for the treatment of a rare disease or condition under
section 527(a) of FDCA, which would run in parallel with the 12
years of data exclusivity of the reference product, if
applicable.
Non-US
regulation
In order to market any product
outside of the US, we would need to comply with numerous and
varying regulatory requirements of other countries and
jurisdictions regarding quality, safety and efficacy, and
governing, among other things, clinical studies, marketing
authorization, commercial sales and distribution of our products.
Whether or not we obtain FDA approval for a product, we would need
to obtain the necessary approvals by the comparable foreign
regulatory authorities before we can commence clinical studies or
marketing of the product in foreign countries and jurisdictions.
Although many of the issues discussed above with respect to the US
apply similarly in the context of the EU, the approval process
varies between countries and jurisdictions and can involve
additional product testing and additional administrative review
periods, as described in greater detail below. The time required to
obtain approval in other countries and jurisdictions might differ
from and be longer than that required to obtain FDA approval.
Regulatory approval in one country or jurisdiction does not ensure
regulatory approval in another, but a failure or delay in obtaining
regulatory approval in one country or jurisdiction may negatively
impact the regulatory process in others.
EU drug
review approval
In the EEA, which is comprised of
the 27 Member States of the EU plus Norway, Iceland and
Liechtenstein medicinal products can only be commercialized after
obtaining a marketing authorization. There are two types of
marketing authorization: the Community Marketing Authorization,
which is issued by the EC through the Centralized Procedure based
on the opinion of the Committee for Medicinal Products for Human
Use (CHMP), a body of the EMA, and which is valid throughout the
entire territory of the EEA; and the National Marketing
Authorization, which is issued by the competent authorities of the
Member States of the EEA and authorizes marketing only in that
Member State’s national territory and not the EEA as a whole.
The Centralized Procedure is
compulsory for human medicines for the treatment of human
immunodeficiency virus or acquired immune deficiency syndrome
(AIDS), cancer, diabetes, neurodegenerative diseases, autoimmune
and other immune dysfunctions, and viral diseases; for veterinary
medicines for use as growth or yield enhancers; for medicines
derived from biotechnology processes, such as genetic engineering;
for advanced-therapy medicines, such as gene-therapy, somatic
cell-therapy or tissue-engineered medicines; and for officially
designated ‘orphan medicines’ (medicines used for rare human
diseases). The Centralized Procedure is optional for products
containing a new active substance not yet authorized in the EEA, or
for products that constitute a significant therapeutic, scientific
or technical innovation, or for products that are in the interest
of public health in the EU. The National Marketing Authorization is
for products not falling within the mandatory scope of the
Centralized Procedure. Where a product has already been authorized
for marketing in a Member State of the EEA, this National Marketing
Authorization can be recognized in another Member State through the
Mutual Recognition Procedure. If the product has not received a
National Marketing Authorization in any Member State at the time of
application, it can be approved simultaneously in various Member
States through the Decentralized Procedure. Under the Decentralized
Procedure, an identical dossier is submitted to the competent
authorities of each of the Member States in which the marketing
authorization is sought, one of which is selected by the applicant
as the Reference Member State (RMS). If the RMS proposes to
authorize the product, and the other Member States do not raise
objections, the product is granted a National Marketing
Authorization in all the Member States in which the authorization
was sought. Before granting the marketing authorization, the EMA or
the competent authorities of the Member States of the EEA assesses
the risk–benefit balance of the product on the basis of scientific
criteria concerning its quality, safety and efficacy.
Regulation in the EU
Product development, the regulatory
approval process, and safety monitoring of medicinal products and
their manufacturers in the EU proceed in much the same manner as
they do in the US. Therefore, many of the issues discussed above
apply similarly in the context of the EU. In addition, drugs are
subject to the extensive price and reimbursement regulations of the
various EU Member States.
Clinical studies
As is the case in the US, the
various phases of preclinical and clinical research in the EU are
subject to significant regulatory controls. The Clinical Trials
Directive 2001/20/EC, as amended and which will be replaced in 2021
or later by Regulation (EU) No 536/2014) provides a system for the
approval of clinical studies in the European Union via
implementation through national legislation of the Member States.
Under this system, approval must be obtained from the competent
national authorities of the EU Member States in which the clinical
trial is to be conducted. Furthermore, a clinical trial may only be
started after a competent ethics committee has issued a favorable
opinion on the clinical trial application, which must be supported
by an investigational medicinal product dossier with supporting
information prescribed by the Clinical Trials Directive and
corresponding national laws of the Member States, and further
detailed in applicable guidance documents. A clinical trial may
only be undertaken if provision has been made for insurance or
indemnity to cover the liability of the investigator or sponsor. In
certain countries, the sponsor of a clinical trial has a strict
(faultless) liability for any (direct or indirect) damage suffered
by trial subjects. The sponsor of a clinical trial, or its legal
representative, must be based in the EEA. European regulators and
ethics committees also require the submission of AE reports during
a study and a copy of the final study report.
Marketing approval
Marketing approvals under the EU
regulatory system may be obtained through a centralized or
decentralized procedure. The centralized procedure results in the
grant of a single marketing authorization, which is valid for all
(currently 27) EU Member States and the three European Free Trade
Association (EFTA) members (Norway, Iceland and
Liechtenstein).
Pursuant to Regulation (EC) No.
726/2004, as amended, the centralized procedure is mandatory for
drugs developed by means of specified biotechnological processes,
advanced-therapy medicinal products, drugs for human use containing
a new active substance for which the therapeutic indication is the
treatment of specified diseases, including but not limited to AIDS,
neurodegenerative disorders, auto-immune diseases and other immune
dysfunctions, as well as drugs designated as orphan drugs. The CHMP
also has the discretion to permit other products to use the
centralized procedure if it considers them sufficiently innovative
or they contain a new active substance.
In the marketing authorization
application, the applicant must properly and sufficiently
demonstrate the quality, safety and efficacy of the drug. Under the
centralized approval procedure, the CHMP, possibly in conjunction
with other committees, is responsible for drawing up the opinion of
the EMA on any matter concerning the admissibility of the files
submitted in accordance with the centralized procedure, such as an
opinion on the granting, variation, suspension or revocation of a
marketing authorization, and pharmacovigilance.
The CHMP and other committees are
also responsible for providing guidelines and have published
numerous guidelines that may apply to our product candidates. These
guidelines provide additional guidance on the factors that the EMA
will consider in relation to the development and evaluation of drug
products and may include, among other things, the preclinical
studies required in specific cases, the manufacturing and control
information that should be submitted in a marketing authorization
application, and the post-approval measures required to monitor
patients and evaluate the long-term efficacy and potential adverse
reactions. Although these guidelines are not legally binding, we
believe that our compliance with them is likely to be necessary to
gain approval for any of our product candidates.
The maximum timeframe for the
evaluation of a marketing authorization application by the CHMP
under the centralized procedure is 210 days after receipt of a
valid application. This period will be suspended until such time as
the supplementary information requested by the CHMP has been
provided by the applicant. Likewise, this time limit will be
suspended for the time allowed for the applicant to prepare oral or
written explanations. When an application is submitted for a
marketing authorization in respect of a drug that is of major
interest from the viewpoint of public health and in particular
therapeutic innovation, the applicant may request an accelerated
assessment procedure. If the CHMP accepts such a request, the time
limit of 210 days will be reduced to 150 days, but it is possible
that the CHMP can revert to the standard time limit for the
centralized procedure if it considers that it is no longer
appropriate to conduct an accelerated assessment.
If the CHMP concludes that the
quality, safety and efficacy of the product are sufficiently
proven, it adopts a positive opinion. This is sent to the EC, which
drafts a decision within approximately 67 days following the CHMP
opinion. After consulting with the Member States, the EC adopts a
decision and grants a marketing authorization, which is valid for
the whole of the EEA. The marketing authorization may be subject to
certain conditions, which may include, without limitation, the
performance of post-authorization safety and/or efficacy
studies.
The EMA has various programs,
including accelerated assessment, conditional approval and PRIority
MEdicines (PRIME), which are intended to increase agency
interactions, expedite or facilitate the process for reviewing
product candidates, and/or provide for initial approval on the
basis of surrogate endpoints. One or more of our product candidates
may qualify for some of these expedited development and review
programs. However, even if a product candidate qualifies for one or
more of these programs, the EMA may later decide that the product
candidate no longer meets the conditions for qualification.
Eligibility to the PRIME scheme is limited to products considered
to offer a major therapeutic advantage in populations with high
unmet need. PRIME is a voluntary scheme aimed at enhancing
interaction and early dialogue with developers of promising
medicines through achieving the early appointment of the Rapporteur
for the product, optimizing development plans and speeding up
evaluation so these medicines can reach patients earlier. Products
benefiting from PRIME can expect to be eligible for accelerated
assessment at the time of application for a marketing authorization
application.
EU legislation also provides for a
system of regulatory data and market exclusivity. According to
Article 14(11) of Regulation (EC) No. 726/2004, as amended, and
Article 10(1) of Directive 2001/83/EC, as amended, upon receiving
marketing authorization, new chemical entities approved on the
basis of a complete independent data package benefit from 8 years
of data exclusivity and an additional 2 years of market
exclusivity. Data exclusivity prevents regulatory authorities in
the EU from referencing the innovator’s data to assess a generic
(abbreviated) application. During the additional 2-year period of
market exclusivity, a generic marketing authorization can be
submitted, and the innovator’s data may be referenced, but no
generic medicinal product can be marketed until the expiration of
the market exclusivity. The overall 10-year period will be extended
to a maximum of 11 years if, during the first 8 years of those 10
years, the marketing authorization holder (MAH) obtains an
authorization for one or more new therapeutic indications that,
during the scientific evaluation prior to their authorization, are
held to bring a significant clinical benefit in comparison with
existing therapies. Even if a compound is considered to be a new
chemical entity and the innovator can gain the period of data
exclusivity, another company nevertheless could also market another
version of the drug if such company obtained marketing
authorization based on a marketing authorization application with a
completely independent data package of pharmaceutical test,
preclinical tests and clinical studies. However, products
designated as orphan medicinal products enjoy, upon receiving
marketing authorization, a period of 10 years of orphan market
exclusivity. See also “—Orphan drug regulation” below. Depending
upon the timing and duration of the EU marketing authorization
process, products may be eligible for an SPC of up to 5 years’,
pursuant to Regulation (EC) No. 469/2009. Such SPCs extend the
rights under the basic patent for the drug.
In the EU, the pediatric regulation
[Regulation (EC) No 1901/2006 as amended] requires sponsors to
submit a pediatric investigation plan at the end of Phase 1. This
plan will provide the details of the quality, non-clinical and
clinical studies required to support the authorization of a
pediatric indication. Additional rules apply to medicinal products
for pediatric use under Regulation (EC) No. 1901/2006. Potential
incentives include a six-month extension of any supplementary
protection certificate granted pursuant to Regulation (EC) No.
469/2009, but not in cases in which the relevant product is
designated as an orphan medicinal product pursuant to Regulation
(EC) No. 141/2000, as amended. Instead, a medicinal product
designated as an orphan medicinal product may enjoy an extension of
the 10-year market exclusivity period granted under Regulation (EC)
No. 141/2000 to 12 years subject to the conditions applicable to
orphan drugs.
Orphan
drug regulation
In the EU, Regulation (EC) No.
141/2000, as amended, states that a drug will be designated as an
orphan drug if its sponsor can establish:
|
• |
that it is intended for the diagnosis, prevention or treatment
of a life-threatening or chronically debilitating condition
affecting not more than 5 in 10,000 persons in the EU when the
application is made, or that it is intended for the diagnosis,
prevention or treatment of a life-threatening, seriously
debilitating or serious and chronic condition in the EU and that
without incentives it is unlikely that the marketing of the drug in
the EU would generate sufficient return to justify the necessary
investment; and
|
|
• |
that there exists no satisfactory method of diagnosis,
prevention or treatment of the condition in question that has been
authorized in the EU or, if such method exists, that the drug will
be of significant benefit to those affected by that
condition.
|
Regulation (EC) No. 847/2000 sets
out further provisions for implementation of the criteria for
designation of a drug as an orphan drug. An application for the
designation of a drug as an orphan drug must be submitted at any
stage of development of the drug before filing of a marketing
authorization application.
If a EU-wide community marketing
authorization in respect of an orphan drug is granted or if all the
EU Member States have granted marketing authorizations in
accordance with the procedures for mutual recognition, the EU and
the Member States will not, for a period of 10 years, accept
another application for a marketing authorization, or grant a
marketing authorization or accept an application to extend an
existing marketing authorization, for the same therapeutic
indication, in respect of a similar drug. This period may, however,
be reduced to 6 years if, at the end of the fifth year, it is
established, with respect to the drug concerned, that the criteria
for orphan-drug designation are no longer met; in other words, when
it is shown on the basis of available evidence that the product is
sufficiently profitable not to justify maintenance of market
exclusivity. Notwithstanding the foregoing, a marketing
authorization may be granted, for the same therapeutic indication,
to a similar drug if:
|
• |
the holder of the marketing authorization for the original
orphan drug has given its consent to the second applicant;
|
|
• |
the holder of the marketing authorization for the original
orphan drug is unable to supply sufficient quantities of the drug;
or
|
|
• |
the second applicant can establish in the application that the
second drug, although similar to the orphan drug already
authorized, is safer, more effective or otherwise clinically
superior.
|
Other incentives available to
orphan drugs in the EU include financial incentives such as a
reduction of fees or fee waivers and protocol assistance.
Orphan-drug designation does not shorten the duration of the
regulatory review and approval process.
Manufacturing and manufacturers’ license
Pursuant to Directive 2003/94/EC,
as transposed into the national laws of the Member States, the
manufacturing of investigational medicinal products and approved
drugs is subject to a separate manufacturer’s license and must be
conducted in strict compliance with cGMP requirements, which
mandate the methods, facilities and controls used in manufacturing,
processing and packing of drugs to assure their safety and
identity. Manufacturers must have at least one qualified person
permanently and continuously at their disposal. The qualified
person is ultimately responsible for certifying that each batch of
finished product released onto the market has been manufactured in
accordance with cGMP and the specifications set out in the
marketing authorization or investigational medicinal product
dossier. cGMP requirements are enforced through mandatory
registration of facilities and inspections of those facilities.
Failure to comply with these requirements could interrupt supply
and result in delays, unanticipated costs and lost revenues, and
subject the applicant to potential legal or regulatory action,
including but not limited to warning letters, suspension of
manufacturing, seizure of product, injunctive action, or possible
civil and criminal penalties.
Wholesale distribution and license
Pursuant to Directive 2001/83/EC,
the wholesale distribution of medicinal products is subject to the
possession of an authorization to engage in activity as a
wholesaler in medicinal products. Possession of a manufacturing
authorization includes authorization to distribute by wholesale the
medicinal products covered by that authorization. The distribution
of medicinal products must comply with the principles and
guidelines of cGDP.
Advertising
In the EU, the promotion of
prescription medicines is subject to intense regulation and
control, including EU and national legislation as well as
self-regulatory codes (industry codes). Advertising legislation
inter alia includes a prohibition on
direct-to-consumer advertising. All advertising of prescription
medicines must be consistent with the product’s approved Summary of
Product Characteristics, and must be factual, accurate, balanced
and not misleading. Advertising of prescription medicines
pre-approval or off-label is not allowed. Some jurisdictions
require that all promotional materials for prescription medicines
be subjected to prior review and approval, either internal or
regulatory.
Other
regulatory requirements
An MAH for a medicinal product is
legally obliged to fulfill a number of obligations by virtue of its
status as an MAH. The MAH can delegate the performance of related
tasks to third parties, such as distributors or marketing partners,
provided that this delegation is appropriately documented and the
MAH maintains legal responsibility and liability.
The obligations
of an MAH include the following:
|
• |
Manufacturing and batch
release—MAHs should guarantee that all manufacturing
operations comply with relevant laws and regulations, applicable
GMPs, and the product specifications and manufacturing conditions
set out in the marketing authorization, and that each batch of
product is subject to appropriate release formalities.
|
|
• |
Availability and continuous
supply—Pursuant to Directive 2001/83/EC, as transposed into
the national laws of the Member States, the MAH for a medicinal
product and the distributors of the said medicinal product actually
placed on the market in a Member State shall, within the limits of
their responsibilities, ensure appropriate and continued supplies
of that medical product to pharmacies and persons authorized to
supply medicinal products so that the needs of patients in the
Member State in question are covered.
|
|
• |
Pharmacovigilance—MAHs are obliged to
establish and maintain a pharmacovigilance system, including a
qualified person responsible for oversight, to submit safety
reports to the regulators and to comply with the good
pharmacovigilance practice guidelines adopted by the EMA.
|
|
• |
Advertising and
promotion—MAHs remain responsible for all advertising and
promotion of their products, including promotional activities by
other companies or individuals on their behalf, and in some cases
must conduct internal or regulatory pre-approval of promotional
materials. Regulation in this area also covers interactions with
healthcare practitioners and/or patient groups, and in some
jurisdictions legal or self-regulatory obligations to disclose such
interactions exist.
|
|
• |
Medical affairs/scientific
service—MAHs are required to disseminate scientific and
medical information on their medicinal products to healthcare
professionals, regulators and patients.
|
|
• |
Legal representation and
distributor issues—MAHs are responsible for regulatory
actions or inactions of their distributors and agents.
|
|
• |
Preparation, filing and
maintenance of the application and subsequent marketing
authorization— MAHs must maintain appropriate records,
comply with the marketing authorization’s terms and conditions,
fulfill reporting obligations to regulators, submit renewal
applications and pay all appropriate fees to the authorities. We
may hold any future marketing authorizations granted for our
product candidates in our own name, or appoint an affiliate or a
collaboration partner to hold marketing authorizations on our
behalf. Any failure by an MAH to comply with these obligations may
result in regulatory action against an MAH and ultimately threaten
our ability to commercialize our products.
|
Pricing and
reimbursement
In the EU, the pricing and
reimbursement mechanisms by private and public health insurers vary
largely by country and even within countries. The public systems
reimbursement for standard drugs is determined by guidelines
established by the legislator or responsible national authority.
The approach taken varies by Member State. Some jurisdictions
operate positive and negative list systems under which products may
only be marketed once a reimbursement price has been agreed. Other
Member States allow companies to fix their own prices for
medicines, but monitor and control company profits and may limit or
restrict reimbursement. The downward pressure on healthcare costs
in general, particularly prescription drugs, has become very
intense. As a result, increasingly high barriers to the entry of
new products are being erected and some EU countries require the
completion of studies that compare the cost-effectiveness of a
particular product candidate with that of currently available
therapies in order to obtain reimbursement or pricing approval.
Special pricing and reimbursement rules may apply to orphan drugs.
Inclusion of orphan drugs in reimbursement systems tend to focus on
the medical usefulness, need, quality and economic benefits to
patients and the healthcare system as for any drug. Acceptance of
any medicinal product for reimbursement may come with cost, use and
often volume restrictions, which again can vary by country. In
addition, results based rules of reimbursement may apply.
Other
US healthcare laws
In addition to FDA restrictions on
marketing of pharmaceutical or biopharmaceutical products, federal
and state healthcare laws restrict certain business practices in
the pharmaceutical and biopharmaceutical industries. These laws
include, but are not limited to, anti-kickback, false claims, data
privacy and security, and transparency statutes and
regulations.
The US federal Anti-Kickback
Statute prohibits, among other things, knowingly and willfully
offering, paying, soliciting or receiving remuneration, directly or
indirectly, to induce, or in return for, purchasing, leasing,
ordering or arranging for the purchase, lease or order of any good,
facility, item or service reimbursable under Medicare, Medicaid or
other federal healthcare programs. The term “remuneration” has been
broadly interpreted to include anything of value, including for
example, gifts, discounts, the furnishing of supplies or equipment,
credit arrangements, payments of cash, waivers of payment,
ownership interests and providing anything at less than its fair
market value. The Anti-Kickback Statute has been interpreted to
apply to arrangements between pharmaceutical and biopharmaceutical
manufacturers on the one hand and prescribers, purchasers and
formulary managers on the other. Although there are a number of
statutory exceptions and regulatory safe harbors protecting certain
common activities from prosecution, the exceptions and safe harbors
are drawn narrowly, and our practices may not in all cases meet all
the criteria for a statutory exception or safe harbor protection.
Practices involving remuneration that may be alleged to be intended
to induce prescribing, purchases or recommendations may be subject
to scrutiny if they do not qualify for an exception or safe harbor.
Failure to meet all the requirements of a particular applicable
statutory exception or regulatory safe harbor does not make the
conduct per se illegal
under the Anti-Kickback Statute. Instead, the legality of the
arrangement will be evaluated on a case-by-case basis based on a
cumulative review of all its facts and circumstances. Several
courts have interpreted the statute’s intent requirement to mean
that if any one purpose of an arrangement involving remuneration is
to induce referrals of federal healthcare-covered business, the
statute has been violated. The Patient Protection and Affordable
Care Act as amended by the Health Care and Education Reconciliation
Act (collectively, the ACA), amended the intent requirement under
the Anti-Kickback Statute and criminal healthcare fraud statutes
(discussed below) such that a person or entity no longer needs to
have actual knowledge of the statute or the specific intent to
violate it in order to have committed a violation. In addition, the
ACA provides that the government may assert that a claim including
items or services resulting from a violation of the federal
Anti-Kickback Statute constitutes a false or fraudulent claim for
purposes of the civil False Claims Act (discussed below). Further,
the Civil Monetary Penalties Law imposes penalties against any
person or entity that, among other things, is determined to have
presented or caused to be presented a claim to a federal health
program that the person knows or should know is for an item or
service that was not provided as claimed or is false or
fraudulent.
The federal false claims laws
prohibit, among other things, any person or entity from knowingly
presenting, or causing to be presented, a false or fraudulent claim
for payment or approval to the federal government or knowingly
making, using or causing to be made or used a false record or
statement material to a false or fraudulent claim to the federal
government. As a result of a modification made by the Fraud
Enforcement and Recovery Act of 2009, a claim includes “any request
or demand” for money or property presented to the US government.
Recently, several pharmaceutical and other healthcare companies
have been prosecuted under these laws for, among other things,
allegedly providing free product to customers with the expectation
that the customers would bill federal programs for the product.
Other companies have been prosecuted for causing false claims to be
submitted because of the companies’ marketing of the product for
unapproved, and thus non-covered, uses. The federal HIPAA created
new federal criminal statutes that prohibit knowingly and willfully
executing, or attempting to execute, a scheme to defraud any
healthcare benefit program, including private third-party payors,
and knowingly and willfully falsifying, concealing or covering up a
material fact or making any materially false, fictitious or
fraudulent statement in connection with the delivery of, or payment
for, healthcare benefits, items or services.
Additionally, the ACA also included
the federal Physician Payments Sunshine Act, which requires that
certain manufacturers of drugs, devices, biologicals and medical
supplies for which payment is available under Medicare, Medicaid or
the Children’s Health Insurance Program (with certain exceptions)
to report information related to certain payments or other
transfers of value made or distributed to physicians and teaching
hospitals, or to entities or individuals at the request of, or
designated on behalf of, the physicians and teaching hospitals and
to report annually certain ownership and investment interests held
by physicians and their immediate family members.
Additionally, many states have
similar healthcare statutes or regulations that apply to items and
services reimbursed under Medicaid and other state programs, or, in
several states, apply regardless of the payor. Certain states
require the posting of information relating to clinical studies,
and require pharmaceutical companies to implement a comprehensive
compliance program that includes a limit on expenditures for, or
payments to, individual medical or health professionals and to
track and report gifts and other payments made to physicians and
other healthcare providers. If our operations are found to be in
violation of any of the health regulatory laws described above or
any other laws that apply to us, we may be subject to penalties,
including potentially significant criminal, civil and/or
administrative penalties, damages, fines, disgorgement, individual
imprisonment, exclusion of products from reimbursement under
government programs, contractual damages, reputational harm,
administrative burdens, diminished profits and future earnings, and
the curtailment or restructuring of our operations, any of which
could adversely affect our ability to operate our business and our
results of operations. To the extent that any of our products will
be sold in a foreign country, we may be subject to similar foreign
laws and regulations, which may include, for instance, applicable
post-marketing requirements, including safety surveillance,
anti-fraud and abuse laws, implementation of corporate compliance
programs and reporting of payments or transfers of value to
healthcare professionals.
Data
privacy and security laws
In addition, we may be subject to
international, federal and state data privacy and security laws,
regulations, rules and standards. Internationally, laws,
regulations and standards in many jurisdictions, such as the GDPR
and the UK GDPR, apply broadly to the collection, use, retention,
security, disclosure, transfer and other processing of personal
information. At the federal level, HIPAA, as amended by the Health
Information Technology for Economic and Clinical Health Act
(HITECH), and its implementing regulations, imposes certain
requirements relating to the privacy, security and transmission of
individually identifiable health information. Among other things,
HITECH makes HIPAA’s privacy and security standards directly
applicable to business associates—independent contractors or agents
of covered entities that receive or obtain protected health
information in connection with providing a service on behalf of a
covered entity. HITECH also created four new tiers of civil
monetary penalties, amended HIPAA to make civil and criminal
penalties directly applicable to business associates, and gave
state attorneys general new authority to file civil actions for
damages or injunctions in federal courts to enforce the federal
HIPAA laws and to seek attorneys’ fees and costs associated with
pursuing federal civil actions. In addition, state laws, (such as
the CCPA and the CPRA,) govern the privacy and security of health
and other personal information in certain circumstances, many of
which differ from each other in significant ways and may not have
the same effect, thus complicating compliance efforts.
Non-compliance with these laws, regulations, rules and standards
could result in significant penalties or legal liability. Although
we take steps to comply with applicable laws, rules and
regulations, we cannot ensure that we will not be subject to
regulatory or private actions, investigations, disputes and
litigation, which may include substantial fines or other legal
liability for noncompliance of data privacy and security laws,
rules and regulations, including in the event of a cybersecurity
breach or other security incident. We could be adversely affected
if legislation or regulations are expanded to require changes in
our or our third-party service providers’ business practices or if
governing jurisdictions interpret or implement their legislation or
regulations in ways that negatively affect our business, results of
operations or financial condition. See “Risk Factors— Changes in laws, rules or regulations
relating to data privacy and security, or any actual or perceived
failure by us to comply with such laws, rules, regulations and
standards, or contractual or other obligations relating to data
privacy and security, could have a material adverse effect on our
reputation, results of operations, financial condition and cash
flows.”
Pharmaceutical
coverage, pricing and reimbursement
In both domestic and foreign
markets, our or our collaboration partners’ sales of any approved
products will depend in part on the availability of coverage and
adequate reimbursement from third-party payors. Third-party payors
include government authorities, managed care providers, private
health insurers and other organizations. Patients who are
prescribed treatments for their conditions and providers performing
the prescribed services generally rely on third-party payors to
reimburse all or part of the associated healthcare costs. Patients
are unlikely to use our products, if approved, unless coverage is
provided and reimbursement is adequate to cover a significant
portion of the cost of our products. Sales of our products will
therefore depend substantially, both domestically and abroad, on
the extent to which the costs of our products will be paid by
third-party payors. These third-party payors are increasingly
focused on containing healthcare costs by challenging the price and
examining the cost-effectiveness of medical products and
services.
In addition, significant
uncertainty exists as to the coverage and reimbursement status of
newly approved healthcare product candidates. The market for our
product candidates for which we may receive regulatory approval
will depend significantly on access to third-party payors’ drug
formularies, or lists of medications for which third-party payors
provide coverage and reimbursement. The industry competition to be
included in such formularies often leads to downward pricing
pressures on pharmaceutical or biopharmaceutical companies.
Additionally, third-party payors may refuse to include a particular
branded drug in their formularies or otherwise restrict patient
access to a branded drug when a less costly generic equivalent or
another alternative is available. Because each third-party payor
individually approves coverage and reimbursement levels, obtaining
coverage and adequate reimbursement is a time-consuming, costly and
sometimes unpredictable process. We may be required to provide
scientific and clinical support for the use of any product to each
third-party payor separately with no assurance that approval would
be obtained, and we may need to conduct expensive pharmacoeconomic
studies in order to demonstrate the cost-effectiveness of our
products. This process could delay the market acceptance of any
product and could have a negative effect on our future revenues and
operating results. We cannot be certain that our product candidates
will be considered cost-effective. Because coverage and
reimbursement determinations are made on a payor-by-payor basis,
obtaining acceptable coverage and reimbursement from one payor does
not guarantee we will obtain similar acceptable coverage or
reimbursement from another payor. If we are unable to obtain
coverage of, and adequate reimbursement and payment levels for, our
product candidates from third-party payors, physicians may limit
how much or under what circumstances they will prescribe or
administer them and patients may decline to purchase them. This in
turn could affect our ability to successfully commercialize our
products and impact our profitability, results of operations,
financial condition and future success.
Furthermore, in many foreign
countries, particularly the countries of the EU, the pricing of
prescription drugs is subject to government control. In some non-US
jurisdictions, the proposed pricing for a drug must be approved
before it may be lawfully marketed. The requirements governing drug
pricing vary widely from country to country. For example, the EU
provides options for its member states to restrict the range of
medicinal products for which their national health insurance
systems provide reimbursement and to control the prices of
medicinal products for human use. A member state may approve a
specific price for the medicinal product or it may instead adopt a
system of direct or indirect controls on the profitability of the
company placing the medicinal product on the market. We may face
competition for our product candidates from lower-priced products
in foreign countries that have placed price controls on
pharmaceutical or biopharmaceutical products. In addition, there
may be importation of foreign products that compete with our own
products, which could negatively impact our profitability.
Healthcare
reform
In the US and other jurisdictions,
there have been, and we expect there will continue to be, a number
of legislative and regulatory changes to the healthcare system that
could affect our future results of operations as we begin to
commercialize our products directly.
In particular, there have been and
continue to be a number of initiatives at the US federal and state
level that seek to reduce healthcare costs. Initiatives to reduce
the federal deficit and to reform healthcare delivery are
increasing cost-containment efforts. We anticipate that Congress,
state legislatures and the private sector will continue to review
and assess alternative benefits, controls on healthcare spending
through limitations on the growth of private health insurance
premiums and Medicare and Medicaid spending, the creation of large
insurance purchasing groups, price controls on pharmaceuticals and
other fundamental changes to the healthcare delivery system. Any
proposed or actual changes could limit or eliminate our spending on
development projects and affect our ultimate profitability.
In March 2010, the ACA, as amended
by the HCERA (collectively, the Health Care Reform Law) was signed
into law. The Health Care Reform Law has the potential to
substantially change the way healthcare is financed by both
governmental and private insurers. The Health Care Reform Law among
other things, established an annual, non-deductible fee on any
entity that manufactures or imports certain branded prescription
drugs and biologic agents; revised the methodology by which rebates
owed by manufacturers for covered outpatient drugs under the
Medicaid Drug Rebate Program are calculated; increased the minimum
Medicaid rebates owed by most manufacturers under the Medicaid Drug
Rebate Program; extended the Medicaid Drug Rebate program to
utilization of certain injectable outpatient drugs, as well as
prescriptions of individuals enrolled in Medicaid managed care
organizations; required manufacturers to offer 50% point-of-sale
discounts on negotiated prices of applicable brand drugs to
eligible beneficiaries during their coverage gap period, as a
condition for the manufacturer’s outpatient drugs to be covered
under Medicare Part D; and implemented payment system reforms
including a national pilot program on payment bundling to encourage
hospitals, physicians and other providers to improve the
coordination, quality and efficiency of certain healthcare services
through bundled payment models. On July 24, 2020 and September 13,
2020, the Trump administration announced several executive orders
related to prescription drug pricing that attempt to implement
several of the administration’s proposals. As a result, the FDA
released a final rule on September 24, 2020, effective November 30,
2020, providing guidance for states to build and submit importation
plans for drugs from Canada. Further, on November 20, 2020, the US
Department of Health and Human Services, or HHS, finalized a
regulation removing safe harbor protection for price reductions
from pharmaceutical manufacturers to plan sponsors under Part D,
either directly or through pharmacy benefit managers, unless the
price reduction is required by law. The rule also creates a new
safe harbor for price reductions reflected at the point-of-sale, as
well as a safe harbor for certain fixed fee arrangements between
pharmacy benefit managers and manufacturers. On November 20, 2020,
CMS issued an interim final rule implementing former President
Trump’s Most Favored Nation executive order, which would tie
Medicare Part B payments for certain physician-administered drugs
to the lowest price paid in other economically advanced countries,
effective January 1, 2021. On December 28, 2020, the US District
Court in Northern California issued a nationwide preliminary
injunction against implementation of the interim final rule. It is
unclear whether the Biden administration will work to reverse these
measures or pursue similar policy initiatives. At the state level,
legislatures have increasingly passed legislation and implemented
regulations designed to control pharmaceutical and biological
product pricing, including price or patient reimbursement
constraints, discounts, restrictions on certain product access and
marketing cost disclosure and transparency measures, and, in some
cases, designed to encourage importation from other countries and
bulk purchasing.
The future of the Health Care
Reform Law remains uncertain. In January 2017, Congress voted to
adopt a budget resolution for the fiscal year 2017 that authorized
the implementation of legislation that would repeal portions of the
Health Care Reform Law. On
December 14, 2018, a federal judge in Texas ruled that the Health
Care Reform Law is unconstitutional in its entirety because the
“individual mandate” was repealed by Congress as part of the 2017
Tax Act. While the judge, as well as the Trump administration and
CMS, have stated that the ruling will have no immediate effect
pending appeal of the decision, it is unclear how this decision,
subsequent appeals, and other efforts to repeal and replace the
ACA, will impact our business. On December 18, 2019, the Fifth
Circuit Court of Appeals upheld the lower court’s decision that the
Health Care Reform Law was unconstitutional. On March 2, 2020, the
US Supreme Court granted certiorari to review the case and oral
arguments were held on November 10, 2020. Although the US Supreme
Court has yet ruled on the constitutionality of the ACA, on January
28, 2021, President Biden issued an executive order to initiate a
special enrollment period from February 15, 2021 through May 15,
2021 for purposes of obtaining health insurance coverage through
the Health Care Reform Law marketplace. The executive order also
instructs certain governmental agencies to review and reconsider
their existing policies and rules that limit access to healthcare,
including among others, reexamining Medicaid demonstration projects
and waiver programs that include work requirements, and policies
that create unnecessary barriers to obtaining access to health
insurance coverage through Medicaid or the ACA. Pending review, the
Health Care Reform Law remains in effect, but it is unclear what
effect this litigation, other efforts to repeal and replace the
Health Care Reform Law and the healthcare reform measures of the
Biden administration will have on the status of the ACA. Litigation
and legislation over the Health Care Reform Law are likely to
continue, with unpredictable and uncertain results.
In the future, there may continue
to be additional proposals relating to the reform of the US
healthcare system, some of which could further limit the prices we
are able to charge for our products candidates, or the amounts of
reimbursement available for our product candidates. If future
legislation were to impose direct governmental price controls and
access restrictions, it could have a significant adverse impact on
our business. Managed care organizations, as well as Medicaid and
other government agencies, continue to seek price discounts. Some
states have implemented, and other states are considering, price
controls or patient access constraints under the Medicaid program,
and some states are considering price-control regimes that would
apply to broader segments of their populations that are not
Medicaid-eligible. Due to the volatility in the current economic
and market dynamics, we are unable to predict the impact of any
unforeseen or unknown legislative, regulatory, payor or policy
actions, which may include cost-containment and healthcare-reform
measures. Such policy actions could have a material adverse impact
on our profitability.
Moreover, the recently enacted
federal Drug Supply Chain Security Act imposes new obligations on
manufacturers of pharmaceutical or biopharmaceutical products,
among others, related to product tracking and tracing. Among the
requirements of this new federal legislation, manufacturers will be
required to provide certain information regarding the drug product
to individuals and entities to which product ownership is
transferred, label drug product with a product identifier, and keep
certain records regarding the drug product. Further, under this new
legislation, manufacturers will have drug product investigation,
quarantine, disposition and notification responsibilities related
to counterfeit, diverted, stolen and intentionally adulterated
products, as well as products that are the subject of fraudulent
transactions or that are otherwise unfit for distribution such that
they would be reasonably likely to result in serious health
consequences or death.
Physician Payment Sunshine Act and transparency
The Physician Payment Sunshine Act
requires most pharmaceutical and biopharmaceutical manufacturers to
report annually to the Secretary of Health and Human Services any
and all financial arrangements, payments, or other transfers of
value made by that entity to physicians and teaching hospitals. The
payment information is made publicly available in a searchable
format on a content management system website. Over the next
several years, we will need to dedicate significant resources to
establish and maintain systems and processes in order to comply
with these regulations. Failure to comply with the reporting
requirements can result in significant civil monetary penalties.
Similar laws have been enacted or are under consideration in
foreign jurisdictions, including France, which has adopted the Loi
Bertrand, or French Sunshine Act, which became effective in 2013.
In addition, the Code of Ethics from the EFPIA requires certain
disclosures of interactions with institutions and healthcare
professionals in various jurisdictions in which we operate.
Environmental,
health and safety laws and regulations
We are subject to numerous
environmental, health and safety laws and regulations and
permitting requirements, including those governing laboratory
procedures, decontamination activities, and the handling,
transportation, use, remediation, storage, treatment, and disposal
of hazardous materials and wastes. Our operations involve the use
of hazardous and flammable materials, and the risk of injury,
contamination or noncompliance with environmental, health and
safety requirements cannot be eliminated. Although compliance with
such laws and regulations and permitting requirements has not had a
material effect on our capital expenditures, earnings or
competitive position, environmental, health and safety laws, and
regulations and permitting requirements have tended to become
increasingly stringent and, to the extent that legal or regulatory
changes may occur in the future, they could result in, among other
things, increased costs to us or the impairment of our research,
development or production efforts.
C.
|
Organizational structure
|
We are a Swiss stock corporation
(société anonyme)
organized under the laws of Switzerland. We were formed as a Swiss
limited liability company (société à responsabilité limitée) on
February 13, 2003 with our registered office and domicile in Basel,
Switzerland. We converted to a Swiss stock corporation
(société anonyme) under
the laws of Switzerland on August 25, 2003. Our Swiss enterprise
identification number is CHE-109.878.825. Prior to our initial
public offering, we were a privately owned company. Our domicile
and registered office is in Ecublens, near Lausanne, Canton of
Vaud, Switzerland. Our registered and principal executive offices
are located at EPFL Innovation Park, Building B, 1015 Lausanne,
Switzerland, our general telephone number is (41) 21 345 91 21 and
our internet address is www.acimmune.com.
The Company
controls a fully-owned subsidiary, AC Immune USA, Inc. (“AC Immune
USA” or “Subsidiary”), which was registered and organized under the
laws of Delaware, USA in Q2 2021. The Company and its Subsidiary
form the Group.
D.
|
Property, plant and equipment
|
The Company’s capital expenditures
were CHF 2.6 million in 2021 with CHF 2.1 million for laboratory
equipment, additional laboratory space and leasehold improvements.
These investments were made to enhance our research
facilities.
Facilities
We do not own any real property.
The table below details the sizes and uses of our leased facilities
as of December 31, 2021:
Location
|
|
Primary Function
|
|
Approximate Size
|
École Polytechnique Fédérale Lausanne (EPFL)
Innovation Park Building B,
1015 Lausanne, Vaud, Switzerland
|
|
Headquarters
Research, discovery, preclinical and clinical
development
Chemistry manufacturing and control
|
|
27,000 square feet
|
1230 Avenue of the Americas
Suite 1634
New York, New York 10020
|
|
US operations
|
|
1,600 square feet
|
The Innovation Park of the EPFL
serves as our corporate headquarters, our research facility and
laboratories. We believe that using the EPFL facilities instead of
building our own infrastructure helps us to maximize the value of
our research and development capital and make efficient use of our
funds as we continue to build and develop our pipeline. We believe
that the space of our existing facilities is sufficient to meet our
current needs.
ITEM 4A.
|
UNRESOLVED STAFF
COMMENTS
|
None.
ITEM 5.
|
OPERATING AND
FINANCIAL REVIEW AND PROSPECTS
|
You should read the following
discussion and analysis of our financial condition and results of
operations together with our audited consolidated financial
statements, including the notes thereto, included in this Annual
Report. The following discussion is based on our financial
information prepared in accordance with IFRS as issued by the IASB,
which might differ in material respects from generally accepted
accounting principles in other jurisdictions. The following
discussion includes forward-looking statements that involve risks,
uncertainties and assumptions. Our actual results may differ
materially from those anticipated in these forward-looking
statements as a result of many factors, including but not limited
to those described under “Item 3. Key information—D. Risk factors”
and elsewhere in this Annual Report.
Overview
To date, we have primarily financed
our operations through the proceeds from our public offerings,
share issuances, contract revenues from license and collaboration
agreements and grants. We have no products approved for
commercialization and have never generated any revenues from
product sales. Pharmaceutical and biopharmaceutical product
development is a highly speculative undertaking and involves a
substantial degree of risk. It may be several years, if ever,
before we or our collaboration partners complete pivotal clinical
studies and have a product candidate approved for
commercialization, and we begin to generate revenue and royalties
from product sales. Since our inception, we have received upfront
and milestone payments from our collaboration partners and certain
other revenue. However, we have also incurred significant operating
losses. We incurred net losses of CHF 73.0 million for the fiscal
year ended December 31, 2021 and have an accumulated losses balance
of CHF 200.9 million as of December 31, 2021.
Strategic collaborations and
licensing agreements
Since our inception, we have
entered into strategic collaboration agreements with a range of
partners covering a number of our product candidates. We entered
into a strategic collaboration with Genentech in November 2006 (as
amended in March 2009, January 2013, May 2014 and May 2015)
regarding the development, manufacture and commercialization of
crenezumab, and we refer to this agreement as the 2006
Agreement.
In June 2012, we entered into an
additional strategic collaboration agreement with Genentech
regarding the development, manufacture and commercialization of
anti-Tau antibodies, which covers semorinemab, and we refer to this
agreement as the 2012 Agreement. We expect to capitalize on
Genentech’s drug development and regulatory expertise and
commercial capabilities to bring our partnered therapeutic products
to market.
In May 2014, we entered into a
license and collaboration agreement with LMI (formerly Piramal
Imaging SA) covering our Tau-PET Imaging tracer.
In December 2014 (as amended in
April 2016, July 2017, January 2019 and November 2019), we entered
into a strategic collaboration agreement with Janssen regarding the
development, manufacture and commercialization of anti-Tau
vaccines, which covers ACI-35. We expect to capitalize on Janssen
and Johnson & Johnson’s extensive regulatory expertise and
experience in developing, manufacturing and, if approved,
commercializing vaccines to bring ACI-35 to market.
We entered into a license agreement
with Lilly in December 2018 (as amended in September 2019 and March
2020) to research and develop Morphomer Tau small molecules for the
treatment of AD and other neurodegenerative diseases. Under the
terms of this agreement, we have completed a Phase 1 clinical study
with ACI-3024. Lilly is
responsible for leading and funding further clinical development
for small molecule Tau aggregation inhibitors with plans to
evaluate candidates in AD and NeuroOrphan indications. Lilly will
also retain global commercialization rights for all
indications.
Genentech, a member of the Roche Group
We have two partnership agreements
with Genentech, a company with a reputation for scientific
excellence and a history of bringing innovative protein
therapeutics to market.
Anti-Abeta
antibody in AD – 2006 agreement
In November 2006, we signed an
exclusive, worldwide licensing agreement for crenezumab, our
humanized monoclonal therapeutic antibody targeting misfolded
Abeta. The agreement was amended March 2009, January 2013, May 2014
and May 2015. The agreement also provides for the development of a
second therapeutic product for a non-AD indication based on the
same intellectual property and anti-Abeta antibody compound. The
value of this partnership is potentially greater than USD 340 (CHF
314) million. The structure of the collaboration agreement is as
follows:
|
• |
A right-of-use license;
|
|
• |
Clinical milestone
payments: payable upon commencement of each of Phase 1 and
Phase 2 of clinical developments, and upon the earlier of
Genentech’s decision to authorize Phase 3 or the commencement of
Phase 3 of clinical developments. In addition, for a second
indication, clinical milestone payments would be payable upon
commencement of Phase 2 of clinical developments and upon the
earlier of Genentech’s decision to authorize Phase 3 or the
commencement of Phase 3 of clinical developments;
|
|
• |
Regulatory milestone
payments: payable upon making regulatory filings in the US
and Europe, respectively, and milestone payments upon obtaining
marketing approval in each of the US and Europe. In addition, for a
second indication, additional regulatory and approval milestones
would be payable.
|
|
• |
Royalties: payable on
sales, with different royalty rates applicable in the US and
Europe. Royalty levels are tied to annual sales volumes.
We may receive royalties on
sales of crenezumab with the percentage rates ranging from the
mid-single digits to mid-teens.
|
To date, we have received total
milestone payments of USD 65 (CHF 70.1) million comprised of an
upfront payment of USD 25 (CHF 31.6) million and of USD 40 (CHF
38.2) million for clinical development milestones achieved, all
prior to January 1, 2017.
Under the terms of the agreement,
Genentech bears all the costs of developing crenezumab through the
clinical phases. In addition, Genentech is responsible for the
costs associated with seeking and obtaining regulatory and
marketing approvals, along with manufacturing sales and marketing
costs. Intellectual property costs related to any
crenezumab-related intellectual property filed solely by us and any
costs associated with filing, maintaining and protecting
intellectual property filed jointly we share with Genentech. The
agreement will terminate by its terms on the date on which all
obligations between the parties with respect to the payment of
milestones or royalties for licensed products have passed or
expired. Either party may terminate the agreement for any material
breach by the other Party, provided a cure period of 90 days from
the date when that notice is given.
On January 30, 2019, we announced
that Roche, the parent of Genentech, is discontinuing the CREAD and
CREAD 2 (BN29552 and BN29553) Phase 3 studies of crenezumab in
people with prodromal-to-mild sporadic AD.
Crenezumab continues to be studied
in the Phase 2 preventive trial, which began in 2013 in Colombia,
of cognitively healthy individuals who carry the PSEN1 E280A
autosomal-dominant mutation and are in a preclinical phase of ADAD.
This study will determine if treating people carrying this mutation
with crenezumab prior to the onset of AD symptoms will slow or
prevent the decline of cognitive and functional abilities. The data
for the primary outcome measures are expected in H1 2022.
Anti-Tau
antibody in AD – 2012 agreement
In June 2012, we entered into a
second agreement with Genentech to research, develop and
commercialize our anti-Tau antibodies for use as immunotherapeutics
and diagnostics. The agreement was amended in December 2015. The
value of this exclusive, worldwide alliance is potentially greater
than CHF 400 million and includes upfront and clinical, regulatory
and commercial milestone payments. In addition to milestones, we
will be eligible to receive royalties on sales at a percentage rate
ranging from the mid-single digits to low-double digits. The
agreement also provides for collaboration on at least one
additional therapeutic indication outside of AD built on the same
anti-Tau antibody program as well an anti-Tau diagnostic product
for AD.
To date, we have received payments
totaling CHF 59 million, including a milestone payment of CHF 14
million received and recognized in Q4 2017 associated with the
first patient dosing in a Phase 2 clinical trial for AD with an
anti-Tau monoclonal body known as semorinemab, a milestone payment
of CHF 14 million recognized in Q2 2016 and received in July 2016,
associated with the announcement of the commencement of the Phase 1
clinical study of semorinemab, and a milestone payment of CHF 14
million received in 2015 in connection with the ED-GO
decision.
The structure of the collaboration
agreement is as follows.
|
• |
A right-of-use license.
|
|
• |
Preclinical and clinical
milestone payments: payable upon selection of a lead
candidate and commencement of each of Phase 1, 2 and 3 of clinical
development. In addition, for a second indication, clinical
milestone payments would be payable upon commencement of each of
Phase 2 and 3 of clinical development.
|
|
• |
Regulatory milestone
payments: payable upon making regulatory filings for
marketing approvals in each of the US, Europe and Japan. In
addition, for a second indication, similar regulatory milestones
would be payable.
|
|
• |
Commercialization
milestones: payable upon making a first commercial sale in
each of the US, Europe and Japan. For a second indication,
commercialization milestones exist for each of the US, Europe and
Japan, which are triggered by the first commercial sale for the
second indication in each of those jurisdictions.
|
|
• |
Royalties: payable on
sales with royalty rates differing based on the source of the
intellectual property underlying the commercial product. We may
receive royalties on sales at a percentage rate ranging from the
mid-single digits to low-double digits
|
Under the terms of the agreement,
Genentech bears all the costs of developing semorinemab through the
clinical phases. In addition, Genentech is responsible for the
costs associated with seeking and obtaining regulatory and
marketing approvals, along with manufacturing, sales and marketing
costs. Intellectual property costs related to any anti-Tau
antibody-related intellectual property filed solely by us and any
costs associated with filing, maintaining and protecting
intellectual property filed jointly we share with Genentech. The
agreement will terminate by its terms on the date on which all
obligations between the parties with respect to the payment of
milestones or royalties for licensed products have passed or
expired. Either party may terminate the agreement for any material
breach by the other Party, provided a cure period of 90 days from
the date when that notice is given.
On September
23, 2020, the Company reported that Genentech informed us of top
line results from a Phase 2 trial of the anti-Tau antibody,
semorinemab, in early (prodromal to mild) Alzheimer’s disease (AD)
which show that semorinemab did not meet its primary efficacy
endpoint of reducing decline on Clinical Dementia Rating-Sum of
Boxes (CDR-SB) compared to placebo. The primary safety endpoint was
however met. Two secondary endpoints, Alzheimer’s Disease
Assessment Scale-Cognitive Subscale 13 (ADAS-Cog13) and Alzheimer’s
Disease Cooperative Study Group – Activities of Daily Living
Inventory (ADCS-ADL), were not met.
On August 31, 2021 the Company
reported that Genentech had informed the Company that the Lauriet
study had met one of its co-primary endpoints, ADAS-Cog 11. The
second co-primary endpoint, ADCS-ADL, was not met. Safety data
showed that semorinemab was well tolerated with an acceptable
safety profile and no unanticipated safety signals. On November 10,
2021, the Company reported that Genentech had presented the full
top-line data from the Lauriet study during a late-breaking session
at the 14th
Clinical Trials on Alzheimer’s Disease conference.
Janssen
Pharmaceuticals, Inc.
Tau Vaccine
in AD – 2014 agreement
In December 2014, we entered into
an agreement with Janssen Pharmaceuticals, Inc. (Janssen) one of
The Janssen Pharmaceutical Companies of Johnson & Johnson, to
develop and commercialize therapeutic anti-Tau vaccines for the
treatment of AD and potentially other Tauopathies. The value of
this collaboration is potentially up to CHF 500 million and
includes upfront and clinical, regulatory and commercial
milestones. In addition to milestones, we will be eligible to
receive royalties on sales at a percentage rate ranging from the
high-single digits to the mid-teens for the phospho-tau vaccine
program. In April 2016, July 2017, January 2019 and November 2019,
the companies entered into the first, second, third and fourth
amendments, respectively. These amendments allow for the alignment
of certain payment and activity provisions with the Development
Plan and Research Plan activities. We and Janssen are co-developing
the second-generation lead therapeutic vaccines, ACI-35.030 and
JACI-35.054, through Phase 1b/2a completion. AC Immune and Janssen
will jointly share research and development costs until the
completion of the first Phase 2b (AC Immune’s contribution to the
first Phase 2b trial is capped). From Phase 2b and onwards, Janssen
will assume responsibility for the clinical development,
manufacturing and commercialization of the second-generation
vaccines.
The Company
received an upfront, non-refundable license fee of CHF 25.9
million, which we recognized as revenue in 2014. In May 2016, we
received a payment of CHF 4.9 million for reaching a clinical
milestone in the Phase 1b study. As we met all performance
obligations on reaching the milestone, we recognized this milestone
as revenue.
The structure of the collaboration
agreement is as follows:
|
• |
A right-of-use license.
|
|
• |
Clinical milestone
payments: payable upon reaching certain milestones in the
Phase 1b study, commencement of the first Phase 2b or 2b/3 of
clinical development, upon reaching enrollment thresholds in the
first Phase 2b or Phase 2b part of the first Phase 2b/3,
commencement of the first Phase 3 or Phase 3 part of a Phase 2b/3
study. In addition, for a second indication, clinical milestone
payments would be payable upon commencement of a Phase 3 clinical
study, which would be payable concurrently with the first
regulatory milestone, if Janssen were to file for regulatory
approval based on Phase 2 clinical data.
|
|
• |
Regulatory milestone
payments: payable upon making regulatory filings in the US,
Europe, and Japan, respectively. In addition, for a second
indication, similar regulatory milestones would be payable. For a
second indication, additional regulatory milestone payments are
payable by Janssen to us upon receipt of each of the regulatory
approvals in the US, Europe and Japan.
|
|
• |
Commercialization
milestones: payable upon making a first commercial sale in
each of the US, Europe and Japan, and upon achieving certain
commercial milestones.
|
|
• |
Royalties: payable on
sales, with royalty rates differing based on the level of annual
sales. We may receive royalties on sales at a percentage rate
ranging from the high-single digits to the mid-teens for the
phospho-tau vaccine program.
|
Under the
terms of the agreement, Janssen may terminate the agreement at any
time after completion of the first Phase 1b clinical study in 2016
by providing 90 days’ notice to us. If not otherwise terminated,
the agreement shall continue until the expiration of all royalty
obligations as outlined in the contract.
LMI
(formerly Piramal Imaging SA)
Tau-PET
imaging agent – 2014 agreement
In May 2014, we entered into an
agreement, our first diagnostic partnership, with LMI, the former
Piramal Imaging SA. The partnership with LMI is an exclusive,
worldwide licensing agreement for the research, development and
commercialization of the Company’s Tau protein PET tracers
supporting the early diagnosis and clinical management of AD and
other Tau-related disorders and includes upfront and sales
milestone payments totaling up to EUR 160 (CHF 167) million, plus
royalties on sales at a percentage rate ranging from mid-single
digits to low-teens. LMI may terminate the LCA at any time by
providing 3 months’ notice to us.
The structure of the collaboration
agreement is as follows:
|
• |
A right-of-use license.
|
|
• |
Clinical milestone
payments: payable upon the commencement of the Phase 1, 2
and 3 studies for generation of data intended to support a
regulatory submission in the US or the EU. We would be entitled to
further clinical milestone payments for the commencement of a Phase
2 and 3 study for a second indication.
|
|
• |
Regulatory milestone
payments: payable upon acceptance of Regulatory filing (NDA)
and Regulatory approval for Commercialization in the US or the
EU.
|
|
• |
Commercialization
milestones: tied to specific annual net sales amounts.
|
|
• |
Royalties: payable on
sales, with royalty rates differing based on the level of annual
sales. We may receive royalties on sales at a percentage rate
ranging from the mid-single digits to the low-teens.
|
Eli
Lilly and Company
Morphomer Tau small molecule – 2018 license agreement
In December 2018, we entered into an
exclusive, worldwide licensing agreement with Eli Lilly and Company
(Lilly) to research and develop Morphomer Tau small molecules for
the treatment of AD and other neurodegenerative diseases.
Per the terms of the agreement,
the Company received an initial upfront payment of CHF 80 million
in Q1 2019 for the rights granted by the Company to Lilly. To date,
the Company has completed a Phase 1 clinical study with
ACI-3024. The program will be expanded to NeuroOrphan
indications and ACI-3024 will be further evaluated for efficacy in
models of rare Tauopathies.
Additionally,
the Company and Lilly have continued candidate characterization
across the research program, identifying new and highly
differentiated candidates with desired cerebrospinal fluid exposure
and selectivity for pathological aggregated Tau. These will be
broadly developed in Tau-dependent neurodegenerative diseases by
Lilly. Lilly is responsible for leading and funding further
clinical development and will retain global commercialization
rights for all indications.
Per the terms of the agreement, the
Company may become eligible to receive additional milestone
payments totaling up to approximately CHF 1.9 billion. In addition to milestones, we will be
eligible to receive royalties on sales at a percentage rate ranging
from the low double-digits to the mid-teens. The agreement
became effective on January 23, 2019 (the “effective date”) when
the waiting period under the Hart-Scott-Rodino Antitrust
Improvements Act of 1976, as amended, expired. In Q3 2019, the Company and Lilly entered
into the first amendment to divide the first discretionary
milestone payment under the agreement of CHF 60 million into two
installments with the first CHF 30 million paid in Q3 2019 and the
second CHF 30 million to be paid on or before March 31, 2020 unless
Lilly terminated the agreement earlier. In Q1 2020, the Company and
Lilly entered into a second amendment to replace the second CHF 30
million to be paid on or before March 31, 2020 with two milestone
payments, one of CHF 10 million to be paid on or before March 31,
2020 and the other of CHF 60 million following the first patient
dosed in a Phase 2 clinical study of a licensed product in the US
or the EU.
The Company
received an initial upfront payment of CHF 80 million in February
2019. We used the residual approach to estimate the selling price
for the right-of-use license and an expected cost plus margin
approach for estimating the research and development activities.
The right-of-use license was delivered on the effective date. The
research and development activities were delivered over time as the
services were performed. For these services, revenue was recognized
over time using the input method, based on costs incurred to
perform the services, as the level of costs incurred over time is
thought to best reflect the transfer of services to Lilly.
The structure of the collaboration
agreement is as follows.
|
• |
An exclusive license: granted by us to
Lilly under certain of our intellectual property to develop,
|
manufacture and commercialize
products containing Morphomer Tau small molecules for the treatment
of AD and other neurodegenerative diseases throughout the world in
any indication.
|
• |
Clinical milestone
payments: payable upon completion of the Lilly preclinical
activities period and following the first patient dosed in a Phase
2 and Phase 3 clinical study of a licensed product in the US or the
EU.
|
|
• |
Regulatory milestone
payments: payable within 60 days after obtaining regulatory
approval for any licensed product in the first indication and any
licensed product in certain additional indications in the US,
Europe and Japan, respectively.
|
|
• |
Commercialization
milestones: payable upon achieving certain
commercial sales milestones.
|
|
• |
Royalties: payable on
sales with royalty rates differing based on the level of annual
sales of licensed products. We may receive royalties on sales at a
percentage rate ranging from the low double-digits to the
mid-teens.
|
The agreement will terminate by the
date of expiration of the last royalty term for the last licensed
product. However, under the terms of the agreement, Lilly may
terminate the agreement at any time after March 31, 2020 by
providing 3 months’ notice to us.
We and Lilly
also entered into a convertible note agreement that became
effective on January 23, 2019 for USD 50 (CHF 50.3) million from
Lilly. In Q2 2019, the Convertible Note Agreement with Lilly
automatically converted in line with the terms of the agreement. As
a result of this conversion, 3,615,328 of our common shares were
issued to Lilly. This note is now fully settled and there is no
further equity or cash consideration due to Lilly thereunder.
Grants
Michael J. Fox Foundation for Parkinson’s Research
In May 2020, the Company, as part
of a joint arrangement with Skåne University Hospital (Skåne) in
Sweden, was awarded a USD 3.2 (CHF 3.0) million grant from the
MJFF’s Ken Griffin Alpha-synuclein Imaging Competition. As part of
this grant, AC Immune is eligible to receive USD 2.5 (CHF 2.3)
million directly from the MJFF. Skåne will receive USD 0.7 (CHF
0.7) million of the total grant directly from the MJFF over two
years to conduct and support the clinical arm of the project.
In December 2021, the Company
announced that it had been awarded two grants totaling USD 1.5 (CHF
1.4) million to advance small molecule PD programs. One award will
support an existing early-stage program to develop small molecules
that can prevent intracellular aggregation and spreading of a-syn.
The other award will fund research on the therapeutic potential of
chemically and mechanistically novel, brain penetrant small
molecule inhibitors of NLRP3 inflammasome activation for the
treatment of PD.
Grant
from the Target ALS Foundation
In Q1 2021, AC Immune was awarded a
USD 0.3 (CHF 0.2) million grant from the Target ALS Foundation
(Target ALS). This grant funds a collaboration between the Company
and the Investigators at the Healey Center for ALS at Massachusetts
General Hospital (MGH) to accelerate the development of the
Company’s proprietary immunoassays to detect disease-associated
forms of TDP-43 in CSF and blood samples.
Critical accounting policies and
significant judgments and estimates
Revenue
recognition
In May 2014,
the IASB issued IFRS 15 Revenue
from Contracts with Customers, which amends the guidance for
accounting for revenues from contracts with customers. This IFRS
replaces all current revenue standards in IFRS including IAS 11
Construction Contracts,
IAS 18 Revenue and various
interpretations.
This standard
applies to all contracts with customers, except for contracts that
are within the scope of other standards, such as leases, insurance,
collaboration arrangements and financial instruments. Under IFRS
15, an entity recognizes revenue when its customer obtains control
of promised goods or services, in an amount that reflects the
consideration that the entity expects to receive in exchange for
those goods or services. To determine revenue recognition for
arrangements that an entity determines are within the scope of IFRS
15, the entity performs the following five steps: (i) identify the
contract(s) with a customer; (ii) identify the performance
obligations in the contract; (iii) determine the transaction price;
(iv) allocate the transaction price to the performance obligations
in the contract; and (v) recognize revenue when (or as) the entity
satisfies a performance obligation. The Company applies the
five-step model to contracts only when it is probable that the
entity will collect the consideration it is entitled to in exchange
for the goods or services it transfers to the customer. At contract
inception, once the contract is determined to be within the scope
of IFRS 15, the Company assesses the goods or services promised
within each contract, and determines those that are performance
obligations, and assesses whether each promised good or service is
distinct. The Company then recognizes as revenue the amount of the
transaction price that is allocated to the respective performance
obligation when (or as) the performance obligation is
satisfied.
Contract revenue. The Company enters into LCAs,
which are within the scope of IFRS 15, under which it licenses
certain proprietary rights to its product candidates and
intellectual property to third parties. The terms of these
arrangements typically include payment to the Company of one or
more of the following: non-refundable, upfront license fees,
development, regulatory and/or commercial milestone payments,
payments for research and clinical services the Company provides
through either its full-time employees or third-party vendors, and
royalties on net sales of licensed commercialized products
depending on the Company’s intellectual property. Each of these
payments results in license, collaboration and other revenues,
which are classified as contract revenue on the consolidated
statements of income/(loss).
Licenses of intellectual property. If
the license to the Company’s intellectual property is determined to
be distinct from the other performance obligations identified in
the arrangement, the Company recognizes revenues from
non-refundable, upfront fees allocated to the license when the
license is transferred to the customer and the customer is able to
use and benefit from the license. For licenses that are sold in
conjunction with a related service, the Company uses judgment to
assess the nature of the combined performance obligation to
determine whether the combined performance obligation is satisfied
over time or at a point in time. If the performance obligation is
settled over time, the Company determines the appropriate method of
measuring progress for purposes of recognizing revenue from
non-refundable, upfront fees. The Company evaluates the measure of
progress each reporting period and, if necessary, adjusts the
measure of performance and related revenue recognition.
Milestone payments. At the inception
of each arrangement that includes development, regulatory and/or
commercial milestone payments, the Company evaluates whether the
milestones are considered highly probable of being reached and
estimates the amount to be included in the transaction price using
the most likely amount method. If it is highly probable that a
significant revenue reversal would not occur in future periods, the
associated milestone value is included in the transaction price.
These amounts for the performance obligations under the contract
are recognized as they are satisfied. At the end of each subsequent
reporting period, the Company re-evaluates the probability of
achievement of such milestones and any related constraint, and if
necessary, adjusts its estimate of the overall transaction price.
Any such adjustments recorded would affect contract revenues and
earnings in the period of adjustment.
Research and development services. The
Company has certain arrangements with our collaboration partners
that include contracting our employees for research and development
programs. The Company assesses if these services are considered
distinct in the context of each contract and, if so, they are
accounted for as separate performance obligations. These revenues
are recorded in contract revenue as the services are
performed.
Sublicense
revenues. The
Company has certain arrangements with our collaboration partners
that include provisions for sublicensing. The Company recognizes
any sublicense revenues at the point in time it is highly probable
to obtain and not subject to reversal in the future.
Contract balances: The Company
receives payments and determines credit terms from its customers
for its various performance obligations based on billing schedules
established in each contract. The timing of revenue recognition,
billings and cash collections results in billed other current
receivables, accrued income (contract assets), and deferred income
(contract liabilities) on the consolidated balance sheets. Amounts
are recorded as other current receivables when the Company’s right
to consideration is unconditional. The Company does not assess
whether a contract has a significant financing component if the
expectation at contract inception is such that the period between
payment by the licensees and the transfer of the promised goods or
services to the licensees will be 1 year or less.
Accrued research and development costs
We record accrued expenses for
estimated costs of our research and development activities
conducted by third-party service providers, which include among
others the conduct of preclinical studies and clinical studies and
contract manufacturing activities. We record accrued expenses for
estimated costs of our research and development activities based
upon the estimated amount of services provided but not yet
invoiced, and we include these costs in accrued expenses on the
consolidated balance sheets and within research and development
expenses in the consolidated statements of income/(loss). These
costs are a significant component of our research and development
expenses.
We record accrued expenses for
these costs based on the estimated amount of work completed in
accordance with agreements established with these third parties,
which involves the following process:
|
• |
communicating with our applicable personnel to identify
services that have been performed on our behalf and estimating the
level of service performed and the associated cost incurred for the
service when we have not yet been invoiced or otherwise notified of
actual costs;
|
|
• |
estimating and accruing expenses in our consolidated financial
statements as of each balance sheet date based on facts and
circumstances known to us at the time; and
|
|
• |
periodically confirming the accuracy of our estimates with
selected providers and adjusting, if necessary.
|
Examples of estimated research
and development expenses that we accrue include:
|
• |
fees paid to CROs in connection with preclinical and
toxicology studies and clinical studies;
|
|
• |
fees paid to investigative sites in connection with clinical
studies;
|
|
• |
fees paid to CMOs in connection with the production of our
product candidates prior to qualifying for capitalization as
inventory; and
|
|
• |
professional service fees for consulting and related
services.
|
We base our
expense accruals related to clinical studies on our estimates of
the services received and efforts expended pursuant to contracts
with multiple research institutions and clinical CROs that conduct
and manage clinical studies on our behalf. The financial terms of
these agreements vary from contract to contract and may result in
uneven payment flows. Payments under some of these contracts depend
on factors such as the successful enrollment of patients and the
completion of clinical study milestones. Our service providers
invoice us monthly in arrears for services performed. In accruing
service fees, we estimate the time period over which the services
will be performed and the level of effort to be expended in each
period. If we do not identify costs that we have begun to incur or
if we underestimate or overestimate the level of services performed
or the costs of these services, our actual expenses could differ
from our estimates.
To date, we
have not experienced significant changes in our estimates of
accrued research and development expenses after a reporting period.
However, due to the nature of estimates, we may be required to make
changes to our estimates in the future as we become aware of
additional information about the status or conduct of our clinical
studies and other research activities.
Share-based compensation
Options
The Company operates an
equity-settled, share-based compensation plan. The fair value of
the employee services received in exchange for the grant of
equity-based awards is recognized as an expense. The total amount
to be expensed over the vesting period is determined by reference
to the fair value of the instruments granted, excluding the impact
of any non-market vesting conditions. Non-market vesting conditions
are included in assumptions about the number of instruments that
are expected to become exercisable. At each balance sheet date, the
Company revises its estimates of the number of instruments that are
expected to become exercisable. It recognizes the impact of the
revision of original estimates, if any, prospectively in the
consolidated statements of income/(loss), and a corresponding
adjustment to equity over the remaining vesting period.
We estimate the fair value of all
time-vested options as of the date of grant using the Black-Scholes
option-pricing model. Key assumptions in determining the
fair value of share options granted utilizing the Black-Scholes
valuation method include the following:
|
Assumption
|
|
Method of estimation
|
|
|
|
|
●
|
Estimated expected term of
options
|
●
|
Simplified method
|
●
|
Expected volatility
|
●
|
Estimate based on average
historical volatilities of common shares of comparable publicly
traded companies. We will continue to apply this process to grants
made as a public company until a sufficient amount of historical
information regarding the volatility of our own stock price becomes
available
|
●
|
Risk-free interest rate
|
●
|
Yields of long-dated Swiss
government zero coupon bond issues
|
●
|
Expected dividends
|
●
|
Zero percent as dividends have
not been paid
|
●
|
Forfeiture rates
|
●
|
Historical and expected
forfeiture data
|
Historically, for all periods prior
to the initial public offering (IPO), the fair value of the common
shares underlying our share-based awards was estimated on each
grant date by our management and approved by our board of
directors. In order to determine the fair value of our common
shares underlying option grants, our board of directors considered,
among other things, the breadth of our product candidate portfolio,
the stages of development of our various product candidates and
major changes to stage of development, the progress and additions
to our collaboration agreements, risks inherent in our activities,
the lack of liquidity of our Company’s securities, and the
valuations and sentiment toward biotech companies. Given the
absence of a public trading market for our common shares, our board
of directors exercised reasonable judgment and considered a number
of objective and subjective factors to determine the best estimate
of the fair value of our common shares, including our stage of
development, progress of our research and development efforts, the
strength of our consolidated balance sheets and capital base,
equity market conditions affecting comparable public companies, and
the lack of liquidity of our common shares.
Restricted
share units
We estimate the fair value of
restricted share units using a reasonable estimate of market value
of the common shares on the date of the award. We classify our
share-based payments as equity-classified awards as they are
settled in our common shares. We measure equity-classified awards
at their grant date fair value and do not subsequently re-measure
them. Compensation costs related to equity-classified awards are
equal to the fair value of the award at grant date amortized over
the vesting period of the award using the graded method. We
reclassify that portion of vested awards to share premium as the
awards vested.
Right-of-use
assets and lease liabilities
Effective
January 1, 2019, the Company adopted IFRS 16 Leases, which provides a new model for
lessee accounting in which all leases, other than short-term and
low-value leases, are accounted for by the recognition on the
consolidated balance sheet of a right-of-use asset and a lease
liability, and the subsequent amortization of the right-of-use
asset over the earlier of the end of the useful life or the lease
term. The Company applied the modified retrospective approach,
which required the recognition of the cumulative effect of
initially applying IFRS 16 as of January 1, 2019 to accumulated
losses and not restating previous years. As the Company recognized
the right-of-use assets at the amount equal to the lease
liabilities there was no impact to accumulated losses. In
accordance with IFRS 16, the Company (i) does not recognize
right-of-use assets and lease liabilities for leases of low value
(i.e. approximate fair value of USD 5,000). For a complete
discussion of accounting, see “Note 5. Right-of-use assets and
lease liabilities.”
In-process
research and development (IPR&D) asset
The Company’s
acquired IPR&D asset is stated at cost less any impairments.
Our IPR&D asset is subject to impairment testing at least
annually or when there are indications that the carrying value may
not be recoverable until the completion of the development process.
At that point, the capitalized amounts are amortized over their
estimated useful life. The determination of the recoverable amounts
include key estimates which are highly sensitive to, and depend
upon, key assumptions.
The Company
will not capitalize future development costs in respect to this
IPR&D asset until they meet the criteria for capitalization of
research and development costs in accordance with IAS 38
Intangible Assets.
Results of operations
The Covid-19 global pandemic has
impacted various countries in which AC Immune currently operates
clinical trials and business operations. The extent to which
Covid-19 may impact us will depend on future developments, which
are currently uncertain and cannot be predicted with confidence,
such as the duration of the outbreak, the severity of Covid-19, or
the effectiveness of actions to contain and treat Covid-19.
The Company continues to effect its
business continuity plan and adapt as the situation evolves.
Currently, we have resumed normal operations at full capacity, with
minimal disruption to our business. We are continuously assessing
and adapting our working practices and business operations to
ensure compliance with official guidance and orders related to the
pandemic and are working proactively with our partners and other
stakeholders to take steps intended to mitigate and minimize any
negative impact to our research, clinical programs and other
business operations.
The Company does not currently have
or project material impacts to the ongoing key trials.
Additionally, the Company has drug supplies that are expected to be
sufficient to complete ongoing trials as well as additional drug
substance supplies expected to be sufficient to support ongoing
cohorts of clinical trials for a period of at least three to six
months. The Company will refrain from starting new clinical trials
if a minimum of a six-month supply on hand cannot be secured.
Finally, the Company currently does not expect delays to its
clinical trials due to manufacturing or supply-chain issues.
Financial operations overview
Contract Revenue
Given our stage of development, we
have not generated any revenue from product sales. Our contract
revenues to date have been derived primarily from separate license
and collaboration agreements on some of our product candidates in
various stages of preclinical and clinical development.
Our contract revenues have
experienced fluctuations over the past three years as a result of
securing new collaboration agreements, the timing of milestone
achievement and the size of each milestone payment. We expect that
any revenue we generate from our collaboration agreements with each
of Lilly, Genentech, Janssen and LMI and/or from any other current
or future collaboration partners will fluctuate from year to year
as a result of the timing and amount of milestones and other
payments.
Research and development expenses
Research and
development costs are expensed as incurred, and consist of salaries
and benefits, laboratory supplies, materials, intellectual
property, facility and information technology (IT) costs, as well
as fees paid to other non-employees and entities that conduct
certain research and development activities on our behalf and all
other allocated expenses. Amounts incurred in connection with
license and collaboration agreements are also included in research
and development expense. Payments made prior to the receipt of
goods or services to be used in research and development are
capitalized until those goods or services are received.
Clinical trial
costs are a component of research and development expenses. We
accrue and expense clinical trial activities performed by third
parties based upon actual work completed in accordance with
agreements established with clinical CROs and clinical sites. We
determine the actual costs through monitoring patient enrollment
and discussions with internal personnel and external service
providers as to the progress or stage of completion of trials or
services and the agreed-upon fee to be paid for such
services.
Manufacturing
start-up costs are a component of research and development
expenses. Additionally, manufacturing costs incurred after
regulatory approval but in connection with significant changes
and/or enhancements to the approved manufacturing process are
recorded as research and development expenses. We accrue and
expense the manufacturing activities performed by third parties
based upon actual work completed in accordance with agreements
established with contract manufacturers.
Our investment in research and
development activities, including the clinical development of our
product candidates has historically been and is projected to be
more than 75% of our total annual operating costs. Research and
development expenses represent costs incurred to conduct research,
such as the discovery and development of our product candidates, as
well as development of new product candidates from our SupraAntigen
and Morphomer platforms and the development of product candidates
pursuant to our collaboration agreements with Lilly, Genentech,
Janssen and LMI. We recognize all research and development costs as
they are incurred. Clinical study costs, contract manufacturing and
other development costs incurred by third parties are expensed as
the contracted work is performed. At present, most of our research
activities comprise three major areas:
|
• |
focused non-AD NDD
including Parkinson’s disease, ALS and NeuroOrphan
indications; and
|
We expect our research and
development expenses to increase substantially in the future and
expect to fund a broader number of projects, which will impact our
research strategy in four key ways:
(i) we expect to undertake
later-stage research and development of our product candidates and,
if approved, to take some of those product candidates into
commercialization;
(ii) we will allocate more funding
to existing programs to advance the development of these
programs;
(iii) we will increase our research
and development efforts on non-AD indications including
NeuroOrphans and diagnostics; and
(iv) we will initiate a number of
new research initiatives that are complementary to our existing and
planned research initiatives.
We expect that our total future research and
development costs will increase over current levels in line with
our three-pillar strategy that focuses on (i) AD, (ii) focused
non-AD NDD including Parkinson’s disease, ALS and
NeuroOrphan indications and (iii) diagnostics.
General and administrative expenses
General and administrative expenses
include personnel costs, expenses for outside professional services
and all other allocated expenses. Personnel costs consist of
salaries, cash bonuses, benefits and share-based compensation.
Outside professional services consist of legal, accounting and
audit services, IT and other consulting fees. Allocated expenses
consist of certain IT, facilities and depreciation expenses. We
continue to incur additional expenses as a result of operating as a
public company, including expenses related to compliance with the
rules and regulations of the SEC, and those of any national
securities exchange on which our securities are traded (Nasdaq),
additional insurance expenses, investor relations activities and
other administrative and professional services.
Other
operating income/(expense)
Other
operating income/(expense) consists primarily of income associated
with foundation grants such as those from the MJFF or Target
ALS.
Finance Result, net
Financial income and expenses
include bank fees associated with charges levied by banks on
foreign payments, interest income and expense associated with our
cash balances and interest
expense associated with lease liabilities. For the year
ended December 31, 2021, we recognized a gain on the change in fair
value of derivative financial assets associated with two
convertible notes sold to certain Affiris affiliated entities that
did not occur in the comparable prior periods. For the year ended
December 31, 2019, we recorded a gain on the conversion feature of
the convertible loan due to Lilly, incurred effective interest to amortize the host debt
for this convertible loan and accrued interest for a
financing obligation.
Exchange differences consist of
foreign exchange transactions and re-measurement gains and losses
that arise from our cash being held in currency other than Swiss
Francs, certain collaboration agreements such as the collaboration
agreements with Genentech and LMI being denominated in currencies
other than Swiss Francs, and selected purchases, which we effect in
foreign currencies.
Taxation
AC Immune is subject to corporate
Swiss federal, cantonal and communal taxation, respectively, in
Switzerland, Canton of Vaud, Commune of Ecublens, near Lausanne.
We are also subject to taxation
in other jurisdictions in which we operate, in particular, the
United States where our wholly-owned subsidiary is
incorporated.
We are entitled under Swiss laws to
carry forward any losses incurred for a period of 7 years and can
offset our losses carried forward against future taxes. As of
December 31, 2021, we had tax loss carry-forwards totaling CHF
197.2 million. There is no certainty that we will make sufficient
profits to be able to utilize these tax loss carry-forwards in
full.
The effective corporate income tax
rate (federal, cantonal and communal) where we are domiciled is
currently 13.606%.
As of January 1, 2020, the Company
may request for 2020 and future tax years a tax relief of 60%,
which would be applied to income from patents and similar rights at
the communal and cantonal levels. This relief would first require
the reintegration of all expensed and deducted research and
development costs related to the concerned patents and similar
rights for consideration in our taxable results from the prior ten
years. The Company has not currently made any decision to enter
this patent box system. Additionally, a “super-deduction” may be
granted for payroll and other expenses of research and development
of Swiss origins.
However, the aforementioned tax
relief based on the patent box and deductions for research and
development may not exceed 50% of the overall taxable profit before
these tax relief and deductions.
Notwithstanding the corporate
income tax, the corporate capital is taxed at a rate of 0.1305%
(cantonal and communal tax only, as there is no federal tax on
capital).
Value added tax (VAT) is charged on
all qualifying goods and services supplied by VAT-registered
businesses. Rates vary based on category, but the Company applies a
standard rate of 7.7% on the value of the goods or services to all
sales invoices, which is payable to the Swiss tax authorities.
Similarly, VAT paid on purchase invoices is reclaimable from the
Swiss tax authorities.
Results of operations
The numbers below have been derived
from our audited consolidated financial statements included
elsewhere in this Annual Report. The discussion below should be
read along with these consolidated financial statements and it is
qualified in its entirety by reference to them.
Comparison of
the years ended December 31, 2021 and 2020
Contract revenue
For the year ended December 31,
2021, AC Immune generated no contract revenues compared with CHF
15.4 million for the comparable period in 2020. This represents a
decrease of CHF 15.4 million. The following table summarizes our
contract revenues during the years ended December 31, 2021 and
2020:
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Contract
revenue
|
|
|
|
|
|
|
|
|
|
|
|
|
Total revenues
|
|
|
|
|
|
|
|
|
|
|
|
|
Our contract revenues experience
fluctuations as a result of securing new collaboration agreements,
the timing of milestone achievements and the size of each milestone
payment. For the year ended
December 31, 2021, the decrease of CHF 15.4 million in contract
revenues compared to the year ended December 31, 2020 is
predominantly related to:
|
• |
a decrease of CHF 14.3 million in our agreement with Lilly.
The Company recognized a CHF 10 million milestone as well as CHF
4.3 million for R&D activities in 2020; and
|
|
• |
a decrease of CHF 1.1 million in our collaboration with
Janssen.
|
Research and development expenses
Research and
development activities are essential to our business and represent
the majority of our costs incurred. Costs for certain development
activities, such as clinical trials, are recognized based on an
evaluation of the progress to completion of specific tasks using
information from the clinical sites and our vendors. Our
collaboration arrangements have different arrangements to share
costs for the development of our product candidates.
We have
completed our R&D spending in both of our Genentech
collaborations. We and Janssen are co-developing second-generation
therapeutic vaccines, ACI-35.030 and JACI-35.054, through Phase
1b/2a completion. AC Immune and Janssen will jointly share research
and development costs until the completion of the first Phase 2b
(AC Immune’s contribution to the first Phase 2b trial is capped).
From Phase 2b and onwards, Janssen will assume responsibility for
the clinical development, manufacturing and commercialization of
the vaccines. We also expect to incur additional R&D
expenditures associated with the expansion of our Morphomer Tau
program into AD and NeuroOrphan indications.
We also intend
to increase our R&D costs associated with the advancement of
ACI-7104 in Parkinson’s disease and our ACI-24 vaccine program
(i.e. ACI-24 AD and ACI-24 DS) through mid- and late-stage clinical
development.
Finally, we intend to further characterize
our other clinical and preclinical candidates. In addition to
these arrangements and proprietarily held assets, we expect
that our total future R&D costs will increase over current
levels, in line with our three-pillar strategy that focuses on (i)
AD, (ii) focused non-AD NDD including Parkinson’s disease, ALS and
NeuroOrphan indications and (iii) diagnostics.
The table below provides a
breakdown of our research and development costs, including direct
research and development costs, manufacturing costs related to
research and development and other research and development costs
not allocated directly to programs for the periods covered by this
Annual Report. The research and development costs not allocated to
specific programs include employment costs, regulatory, QA and
intellectual property costs. We
do not assign our internal costs, such as salary and benefits,
share-based compensation expenses, laboratory supplies, and other
direct expenses and infrastructure costs to individual R&D
projects, because the employees within our R&D groups are
typically deployed across multiple research and development
programs.
For the year ended December 31,
2021, research and development expenses totaled CHF 62.3 million
compared with CHF 59.5 million for the comparable period in 2020.
This represents an increase of CHF 2.8 million. The following table presents the research
and development expenses during the years ended December 31, 2021
and 2020:
Detailed
research and development expenditures by major development
category
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Discovery
and preclinical expenses
|
|
|
19,963
|
|
|
|
20,408
|
|
|
|
(445
|
)
|
Clinical
expenses
|
|
|
14,872
|
|
|
|
17,124
|
|
|
|
(2,252
|
)
|
Group
function expenses
|
|
|
|
|
|
|
|
|
|
|
|
|
Total Direct R&D
|
|
|
|
|
|
|
|
|
|
|
|
|
Payroll
expenses
|
|
|
16,465
|
|
|
|
14,424
|
|
|
|
2,041
|
|
Share-based
compensation
|
|
|
1,528
|
|
|
|
1,276
|
|
|
|
252
|
|
Other
non-allocated
|
|
|
|
|
|
|
|
|
|
|
|
|
Total R&D
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Operating
expenses1
|
|
|
44,289
|
|
|
|
43,787
|
|
|
|
502
|
|
Salaries
and related costs2
|
|
|
|
|
|
|
|
|
|
|
|
|
Total research and development expenses
|
|
|
|
|
|
|
|
|
|
|
|
|
1Includes
depreciation expenses
2Includes
share-based compensation
For the year ended December 31, 2021:
Discovery and preclinical expenses
decreased CHF 0.4 million, primarily due to:
|
• |
a decrease of CHF 2.0 million for
the research of alpha-synuclein antibodies.
|
partially offset by:
|
• |
an increase of CHF 0.7 million for the expansion of our
Morphomer Tau program into NeuroOrphan indications and the further
characterization of our preclinical candidates, CHF 0.5 million for
the optimization and development of our anti-TDP-43 antibody, CHF
0.1 million for our diagnostic imaging agents and CHF 0.4 million
in other discovery programs,
|
Clinical expenses decreased by CHF
2.3 million, primarily due to:
|
• |
a decrease of CHF 2.2
million for Phase 1 activities for our Morphomer Tau compound which
completed in 2020, CHF 2.0 million for ACI-24 for DS as a result of
prior period scaling up activities for a Phase 2 clinical trial
which were not repeated in the current period and CHF 1.8 million
for ACI-24 for AD as the six-month safety period
completed,
|
partially offset by:
|
• |
an increase of CHF 3.5 million for ACI-35.030 driven by
R&D cost sharing, increased patient enrollment into the Phase
1b/2a study and increased frequency of interim analysis testing and
CHF 0.2 million for other clinical programs.
|
The variances in Group function
expenses relate to regulatory and quality assurance, intellectual
property and other non-allocated costs.
Total salaries and related costs
increased by CHF 2.3 million, primarily due to:
|
• |
an increase in salary and benefit related costs of CHF 2.0
million primarily related to the internal reallocation of certain
employees’ personnel cost from general and administrative expenses
to research and development personnel expenses in 2021 and the
annualization of 2020 hires; and
|
|
• |
higher share-based compensation expense of CHF 0.3 million
related predominantly to an increase of stock options issued to
employees.
|
The CHF 3.2 million increase in
other non-allocated expenses relate to administrative R&D and
certain non-allocated functional expenses, primarily due to:
|
• |
an increase of CHF 2.8 million associated with the
reallocation of certain IT and facilities expenditures made in 2021
that were not reclassified in the prior period, CHF 0.3 million in
depreciation expense and CHF 0.1 million in other items.
|
General and administrative expenses
General and
administrative expenses consist primarily of salaries and related
costs, including share-based compensation, professional fees such
as legal and accounting related services, infrastructure expenses,
and other operating expenses.
For the year ended December 31, 2021,
general and administrative expenses totaled CHF 17.9 million
compared with CHF 18.6 million for the comparable period in 2020.
This represents a decrease of CHF 0.7 million. The following table
presents the general and administrative expenses during the years
ended December 31, 2021 and 2020:
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Operating
expenses1
|
|
|
7,031
|
|
|
|
7,471
|
|
|
|
(440
|
)
|
Salaries
and related costs2
|
|
|
|
|
|
|
|
|
|
|
|
|
Total general and administrative expenses
|
|
|
|
|
|
|
|
|
|
|
|
|
1Includes
depreciation expenses
2Includes
share-based compensation
For the year ended December 31, 2021, this decrease is
primarily due to:
|
• |
CHF 2.8 million associated with the reallocation of certain IT
and facilities expenditures made in 2021 that were not reclassified
in the prior period,
|
partially offset by:
|
• |
a CHF 1.1 million for
transaction costs associated with our asset acquisition for a
portfolio of therapeutics targeting alpha-synuclein from
Affiris,
|
|
• |
a CHF 0.9 million increase in our directors’ and officers’
insurance for the period; and
|
|
• |
CHF 0.3 million increase in other administrative
expenses.
|
Other
operating income/(expense)
For the year ended December 31, 2021, other
operating income/(expense) totaled CHF 1.2 million in income
compared with CHF 1.4 million in income for the comparable period
in 2020. This represents a decrease of CHF 0.2 million. The
following table presents the other operating income/(expense)
during the years ended December 31, 2021 and 2020:
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Other
operating income/(expense)
|
|
|
|
|
|
|
|
|
|
|
|
|
Total other operating income/(expense)
|
|
|
|
|
|
|
|
|
|
|
|
|
For the year ended December 31, 2021, this decrease is
primarily due to:
|
• |
a decrease of CHF 0.3 million in
grant income related to activities completed prior to the start of
the current period related to our MJFF awards in 2021,
|
partially offset by;
|
• |
an increase of CHF 0.1 million in
grant income for activities completed for our Target ALS Foundation
award.
|
Finance result, net
For the year ended December 31, 2021,
finance result was a CHF 6.0 million gain compared with a CHF 0.7
million loss for the comparable period in 2020. This represents an
increase of CHF 6.7 million. The following table presents the
finance result during the years ended December 31, 2021 and
2020:
|
|
For the Years
Ended
December
31,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Financial
income
|
|
|
6,485
|
|
|
|
78
|
|
|
|
6,407
|
|
Financial
expense
|
|
|
(581
|
)
|
|
|
(184
|
)
|
|
|
(397
|
)
|
Exchange
differences
|
|
|
|
|
|
|
|
|
|
|
|
|
Finance result, net
|
|
|
|
|
|
|
|
|
|
|
|
|
Net finance
result was a gain primarily increased related to:
|
• |
an increase of CHF 6.4 million in financial income,
predominantly related to a CHF 6.5 million gain on the change in
fair value of derivative financial assets associated with two
convertible notes sold to certain Affiris affiliated entities as a
result of fair value remeasurements; and
|
|
• |
a CHF 0.7 million increase
in favorable foreign currency exchange differences related to
movement in the CHF versus foreign currencies, predominantly the US
Dollar and Euro,
|
partially offset by:
|
• |
a CHF 0.4 million increase in
financial expense, of which CHF 0.4 million relates to interest
expense as many of our CHF-denominated deposit accounts bear
negative interest as well as our lease liabilities in accordance
with IFRS 16.
|
Non-IFRS financial measures
In addition to our operating
results, as calculated in accordance with IFRS, as adopted by the
IASB, we use adjusted income/(loss) and adjusted earnings/(loss)
per share when monitoring and evaluating our operational
performance. Adjusted income/(loss) is defined as income/(loss) for
the relevant period, as adjusted for certain items that we believe
are not indicative of our ongoing operating performance. Adjusted
earnings/(loss) per share is defined as adjusted income/(loss) for
the relevant period divided by the weighted-average number of
shares for such period.
We believe that these measures
assist our shareholders because they enhance the comparability of
our results each period and provide more useful insight into
operational results for the period. The Company’s executive
management uses these non-IFRS measures to evaluate our operational
performance. These non-IFRS financial measures are not meant to be
considered alone or as substitutes for our IFRS financial measures,
and should be read in conjunction with our consolidated financial
statements prepared in accordance with IFRS. The most directly
comparable IFRS measure to these non-IFRS measures is net
income/(loss). The following table reconciles net income/(loss) to
adjusted income/(loss) and adjusted earnings/(loss) per share for
the periods presented:
Reconciliation
of income/(loss) to adjusted income/(loss) and
earnings/(loss)
per share to adjusted earnings/(loss) per share
|
|
For the Years
Ended
December
31,
|
|
(In CHF
thousands, except for share and per share data)
|
|
|
|
|
|
|
|
|
|
Income/(loss)
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjustments:
|
|
|
|
|
|
|
|
|
|
|
|
|
Non-cash share-based
payments1
|
|
|
4,126
|
|
|
|
4,088
|
|
|
|
2,834
|
|
Foreign currency
(gains)/losses2
|
|
|
70
|
|
|
|
703
|
|
|
|
826
|
|
Change in fair value of derivative
financial assets3
|
|
|
(6,459
|
)
|
|
|
—
|
|
|
|
—
|
|
Transaction costs4
|
|
|
1,144
|
|
|
|
—
|
|
|
|
—
|
|
Effective interest
expenses5
|
|
|
—
|
|
|
|
—
|
|
|
|
1,355
|
|
Change in fair value of conversion
feature6
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjusted income/(loss)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Earnings/(loss) per share –
basic
|
|
|
|
|
|
|
|
|
|
|
|
|
Earnings/(loss) per share –
diluted
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjustment to earnings/(loss) per
share – basic
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjustment to earnings/(loss) per
share – diluted
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjusted earnings/(loss) per share
– basic
|
|
|
|
|
|
|
|
|
|
|
|
|
Adjusted earnings/(loss) per share
– diluted
|
|
|
|
|
|
|
|
|
|
|
|
|
Weighted-average number of shares used to compute adjusted
loss per share – basic
|
|
|
74,951,833
|
|
|
|
71,900,212
|
|
|
|
70,603,611
|
|
Weighted-average number of shares used to compute adjusted loss per
share – diluted
|
|
|
74,951,833
|
|
|
|
|